1
|
Quiñonez-Bastidas GN, Grijalva-Contreras LE, Patiño-Camacho SI, Navarrete A. Emerging Psychotropic Drug for the Treatment of Trigeminal Pain: Salvinorin A. Pharmaceuticals (Basel) 2024; 17:1619. [PMID: 39770461 PMCID: PMC11728561 DOI: 10.3390/ph17121619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Trigeminal neuralgia (TN) is chronic pain caused by damage to the somatosensorial system on the trigeminal nerve or its branches, which involves peripheral and central dysfunction pain pathways. Trigeminal pain triggers disruptive pain in regions of the face, including within and around the mouth. Besides clinical experiences, translating the language of suffering into scientific terminology presents substantial challenges. Due to the complex and multifactorial pathophysiology underlying trigeminal pain, elucidating its social impact presents significant difficulties. Carbamazepine and oxcarbazepine are first-line treatments for TN, achieving approximately 50% pain reduction in 60-70% of treated patients. However, their efficacy is often limited by common side effects, such as dizziness, vertigo, nausea, seizures, and cognitive symptoms. In some cases, patients experience severe side effects, including myelosuppression, hyponatremia, hormonal imbalances, liver toxicity, suicidal ideation, teratogenicity, and other adverse reactions. Given these clinical limitations, the search for new painkiller candidates continues. Hence, we focused this review on salvinorin A (SalA), a natural agonist of κ-opioid receptors (KORs), which demonstrated anti-nociceptive, anti-inflammatory, and anti-neuropathic properties in various experimental models of the spinal sensory system. Furthermore, preclinical evidence indicates that SalA does not induce dependence and demonstrates a favorable toxicological and safety profile in comparison with currently marketed opioid drugs. We propose Salvinorin A as a promising candidate for treating trigeminal neuralgia, offering the potential for reduced adverse effects.
Collapse
Affiliation(s)
- Geovanna Nallely Quiñonez-Bastidas
- Centro de Investigación y Docencia en Ciencias de la Salud, Universidad Autónoma de Sinaloa, Eustaquio Buelna 91, Burócrata, Culiacan 80030, Mexico
| | - Lucia Elhy Grijalva-Contreras
- Programa de Licenciatura en Fisioterapia, Universidad Estatal de Sonora, Unidad Académica Hermosillo, Hermosillo 83100, Mexico;
| | - Selene Isabel Patiño-Camacho
- Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Ciudad Universitaria, Culiacan 80013, Mexico;
| | - Andrés Navarrete
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de Mexico 04510, Mexico;
| |
Collapse
|
2
|
Lu J, Zhang L, Zhang J, Sun Y, Wang H, Wang W, Wang K, Qin L, Jia J. Oxidative stress plays an important role in the central regulatory mechanism of orofacial hyperalgesia under low estrogen conditions. Behav Brain Res 2024; 469:115047. [PMID: 38759799 DOI: 10.1016/j.bbr.2024.115047] [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: 02/05/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
Hyperalgesia occurs in the orofacial region of rats when estrogen levels are low, although the specific mechanism needs to be investigated further. Furthermore, oxidative stress plays an important role in the transmission of pain signals. This study aimed to explore the role of oxidative stress in orofacial hyperalgesia under low estrogen conditions. We firstly found an imbalance between oxidative and antioxidant capacity within the spinal trigeminal subnucleus caudalis (SP5C) of rats after ovariectomy (OVX), resulting in oxidative stress and then a decrease in the orofacial pain threshold. To investigate the mechanism by which oxidative stress occurs, we used virus as a tool to silence or overexpress the excitatory amino acid transporter 3 (EAAT3) gene. Further investigation revealed that the regulation of glutathione (GSH) and reactive oxygen species (ROS) can be achieved by regulating EAAT3, which in turn impacts the occurrence of oxidative stress. In summary, our findings suggest that reduced expression of EAAT3 within the SP5C of rats in the low estrogen state may decrease GSH content and increase ROS levels, resulting in oxidative stress and ultimately lead to orofacial hyperalgesia. This suggests that antioxidants could be a potential therapeutic direction for orofacial hyperalgesia under low estrogen conditions, though more research is needed to understand its mechanism.
Collapse
Affiliation(s)
- Jiali Lu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Linqian Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Jinglin Zhang
- Yuncheng Vocational Nursing College, Yuncheng, China
| | - Yanrong Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hanfei Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wenjuan Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ke Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lihua Qin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| | - Jing Jia
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China; Department of Stomatology, The Third Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, China.
| |
Collapse
|
3
|
Cramer N, Ji Y, Kane MA, Pilli NR, Castro A, Posa L, Van Patten G, Masri R, Keller A. Elevated Serotonin in Mouse Spinal Dorsal Horn Is Pronociceptive. eNeuro 2023; 10:ENEURO.0293-23.2023. [PMID: 37945351 PMCID: PMC10698626 DOI: 10.1523/eneuro.0293-23.2023] [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: 08/05/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Serotonergic neurons in the rostral ventral medulla (RVM) contribute to bidirectional control of pain through modulation of spinal and trigeminal nociceptive networks. Deficits in this pathway are believed to contribute to pathologic pain states, but whether changes in serotonergic mechanisms are pro- or antinociceptive is debated. We used a combination of optogenetics and fiber photometry to examine these mechanisms more closely. We find that optogenetic activation of RVM serotonergic afferents in the spinal cord of naive mice produces mechanical hypersensitivity and conditioned place aversion (CPA). Neuropathic pain, produced by chronic constriction injury of the infraorbital nerve (CCI-ION), evoked a tonic increase in serotonin (5HT) concentrations within the spinal trigeminal nucleus caudalis (SpVc), measured with liquid chromatography-tandem mass spectroscopy (LC-MS/MS). By contract, CCI-ION had no effect on the phasic serotonin transients in SpVc, evoked by noxious pinch, and measured with fiber photometry of a serotonin sensor. These findings suggest that serotonin release in the spinal cord is pronociceptive and that an increase in sustained serotonin signaling, rather than phasic or event driven increases, potentiate nociception in models of chronic pain.
Collapse
Affiliation(s)
- Nathan Cramer
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
- University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Yadong Ji
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201
| | - Nageswara R Pilli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201
| | - Alberto Castro
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Luca Posa
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Gabrielle Van Patten
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Radi Masri
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201
- University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Asaf Keller
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201
- University of Maryland - Medicine Institute for Neuroscience Discovery, University of Maryland School of Medicine, Baltimore, MD 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
4
|
Tao R, Liu S, Crawford J, Tao F. Gut-Brain Crosstalk and the Central Mechanisms of Orofacial Pain. Brain Sci 2023; 13:1456. [PMID: 37891825 PMCID: PMC10605055 DOI: 10.3390/brainsci13101456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/21/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Accumulated evidence has demonstrated that the gut microbiome can contribute to pain modulation through the microbiome-gut-brain axis. Various relevant microbiome metabolites in the gut are involved in the regulation of pain signaling in the central nervous system. In this review, we summarize recent advances in gut-brain interactions by which the microbiome metabolites modulate pain, with a focus on orofacial pain, and we further discuss the role of gut-brain crosstalk in the central mechanisms of orofacial pain whereby the gut microbiome modulates orofacial pain via the vagus nerve-mediated direct pathway and the gut metabolites/molecules-mediated indirect pathway. The direct and indirect pathways both contribute to the central regulation of orofacial pain through different brain structures (such as the nucleus tractus solitarius and the parabrachial nucleus) and signaling transmission across the blood-brain barrier, respectively. Understanding the gut microbiome-regulated pain mechanisms in the brain could help us to develop non-opioid novel therapies for orofacial pain.
Collapse
Affiliation(s)
| | | | | | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, 3302 Gaston Ave., Dallas, TX 75246, USA
| |
Collapse
|
5
|
Cramer N, Ji Y, Kane M, Pilli N, Posa L, Patten GV, Masri R, Keller A. Elevated serotonin in mouse spinal dorsal horn is pronociceptive. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552838. [PMID: 37645759 PMCID: PMC10461991 DOI: 10.1101/2023.08.10.552838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Serotonergic neurons in the rostral ventral medulla (RVM) contribute to bidirectional control of pain through modulation of spinal and trigeminal nociceptive networks. Deficits in this pathway are believed to contribute to pathological pain states, but whether changes in serotonergic mechanisms are pro or anti-nociceptive are debated. We used a combination of optogenetics and fiber photometry to examine these mechanisms more closely. We find that optogenetic activation of RVM serotonergic afferents in the spinal cord of naïve mice produces mechanical hypersensitivity and conditioned place aversion. Neuropathic pain, produced by chronic constriction injury of the infraorbital nerve (CCI-ION), evoked a tonic increase in serotonin concentrations within the spinal trigeminal nucleus caudalis (SpVc), measured with liquid chromatography-tandem mass spectroscopy (LC-MS/MS). By contract, CCI-ION had no effect on the phasic serotonin transients in SpVc, evoked by noxious pinch, and measured with fiber photometry of a serotonin sensor. These findings suggest that serotonin release in the spinal cord is pronociceptive and that an increase is sustained serotonin signaling, rather than phasic or event driven increases, potentiate nociception in models of chronic pain.
Collapse
Affiliation(s)
- Nathan Cramer
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yadong Ji
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, Maryland 21201
| | - Maureen Kane
- Department of Pharmaceutical Sciences University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Nageswara Pilli
- Department of Pharmaceutical Sciences University of Maryland School of Pharmacy, Baltimore, Maryland 21201
| | - Luca Posa
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Gabrielle Van Patten
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Radi Masri
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, Maryland 21201
- UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Asaf Keller
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
6
|
Smith JA, Ji Y, Lorsung R, Breault MS, Koenig J, Cramer N, Masri R, Keller A. Parabrachial Nucleus Activity in Nociception and Pain in Awake Mice. J Neurosci 2023; 43:5656-5667. [PMID: 37451980 PMCID: PMC10401640 DOI: 10.1523/jneurosci.0587-23.2023] [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: 03/29/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
The parabrachial nuclear complex (PBN) is a nexus for aversion and for the sensory and affective components of pain perception. We have previously shown that during chronic pain PBN neurons in anesthetized rodents have amplified activity. We report a method to record from PBN neurons of behaving, head-restrained mice while applying reproducible noxious stimuli. We find that both spontaneous and evoked activity are higher in awake animals compared with urethane anesthetized mice. Fiber photometry of calcium responses from calcitonin-gene-related peptide-expressing PBN neurons demonstrates that these neurons respond to noxious stimuli. In both males and females with neuropathic or inflammatory pain, responses of PBN neurons remain amplified for at least 5 weeks, in parallel with increased pain metrics. We also show that PBN neurons can be rapidly conditioned to respond to innocuous stimuli after pairing with noxious stimuli. Finally, we demonstrate that changes in PBN neuronal activity are correlated with changes in arousal, measured as changes in pupil area.SIGNIFICANCE STATEMENT The parabrachial complex is a nexus of aversion, including pain. We report a method to record from parabrachial nucleus neurons of behaving mice while applying reproducible noxious stimuli. This allowed us to track parabrachial activity over time in animals with neuropathic or inflammatory pain. It also allowed us to show that the activity of these neurons correlates with arousal states and that these neurons can be conditioned to respond to innocuous stimuli.
Collapse
Affiliation(s)
- Jesse A Smith
- Program in Neuroscience, Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Yadong Ji
- Department of Advanced Oral Sciences and Therapeutics, School of Dentistry, University of Maryland, Baltimore, Maryland 21201-1786
| | - Rebecca Lorsung
- Program in Neuroscience, Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Macauley S Breault
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Jeffrey Koenig
- Program in Molecular Medicine, Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Nathan Cramer
- Program in Neuroscience, Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Radi Masri
- Program in Neuroscience, Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Department of Advanced Oral Sciences and Therapeutics, School of Dentistry, University of Maryland, Baltimore, Maryland 21201-1786
| | - Asaf Keller
- Program in Neuroscience, Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
7
|
Mo SY, Xue Y, Li Y, Zhang YJ, Xu XX, Fu KY, Sessle BJ, Xie QF, Cao Y. Descending serotonergic modulation from rostral ventromedial medulla to spinal trigeminal nucleus is involved in experimental occlusal interference-induced chronic orofacial hyperalgesia. J Headache Pain 2023; 24:50. [PMID: 37165344 PMCID: PMC10173589 DOI: 10.1186/s10194-023-01584-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/21/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Dental treatment associated with unadaptable occlusal alteration can cause chronic primary myofascial orofacial pain. The serotonin (5-HT) pathway from the rostral ventromedial medulla (RVM) exerts descending modulation on nociceptive transmission in the spinal trigeminal nucleus (Sp5) and facilitates chronic pain. The aim of this study was to investigate whether descending 5-HT modulation from the RVM to the Sp5 is involved in the maintenance of primary myofascial orofacial hyperalgesia after persistent experimental occlusal interference (PEOI) or after delayed removal of experimental occlusal interference (REOI). METHODS Expressions of 5-HT3A and 5-HT3B receptor subtypes in the Sp5 were assessed by immunofluorescence staining and Western blotting. The release and metabolism of 5-HT in the Sp5 were measured by high-performance liquid chromatography. Changes in the pain behavior of these rats were examined after specific pharmacologic antagonism of the 5-HT3 receptor, chemogenetic manipulation of the RVM 5-HT neurons, or selective down-regulation of 5-HT synthesis in the RVM. RESULTS Upregulation of the 5-HT3B receptor subtype in the Sp5 was found in REOI and PEOI rats. The concentration of 5-HT in Sp5 increased significantly only in REOI rats. Intrathecal administration of Y-25130 (a selective 5-HT3 receptor antagonist) dose-dependently reversed the hyperalgesia in REOI rats but only transiently reversed the hyperalgesia in PEOI rats. Chemogenetic inhibition of the RVM 5-HT neurons reversed the hyperalgesia in REOI rats; selective down-regulation of 5-HT in advance also prevented the development of hyperalgesia in REOI rats; the above two manipulations did not affect the hyperalgesia in PEOI rats. However, chemogenetic activation of the RVM 5-HT neurons exacerbated the hyperalgesia both in REOI and PEOI rats. CONCLUSIONS These results provide several lines of evidence that the descending pathway from 5-HT neurons in the RVM to 5-HT3 receptors in the Sp5, plays an important role in facilitating the maintained orofacial hyperalgesia after delayed EOI removal, but has a limited role in that after persistent EOI.
Collapse
Affiliation(s)
- Si-Yi Mo
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Yang Xue
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Yuan Li
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Yao-Jun Zhang
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Xiao-Xiang Xu
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China.
| | - Kai-Yuan Fu
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
- Center for Temporomandibular Disorders and Orofacial Pain, School and Hospital of Stomatology, Peking University, Beijing, 100081, PR China
| | - Barry J Sessle
- Faculty of Dentistry & Department of Physiology, Temerty Faculty of Medicine & Centre for the Study of Pain, University of Toronto, Toronto, ON, M5G 1G6, Canada
| | - Qiu-Fei Xie
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Ye Cao
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
- National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of the People's Republic of China, Peking University, Beijing, 100083, PR China.
| |
Collapse
|
8
|
Cooper AH, Hedden NS, Prasoon P, Qi Y, Taylor BK. Postsurgical Latent Pain Sensitization Is Driven by Descending Serotonergic Facilitation and Masked by µ-Opioid Receptor Constitutive Activity in the Rostral Ventromedial Medulla. J Neurosci 2022; 42:5870-5881. [PMID: 35701159 PMCID: PMC9337598 DOI: 10.1523/jneurosci.2038-21.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 01/29/2023] Open
Abstract
Following tissue injury, latent sensitization (LS) of nociceptive signaling can persist indefinitely, kept in remission by compensatory µ-opioid receptor constitutive activity (MORCA) in the dorsal horn of the spinal cord. To demonstrate LS, we conducted plantar incision in mice and then waited 3-4 weeks for hypersensitivity to resolve. At this time (remission), systemic administration of the opioid receptor antagonist/inverse agonist naltrexone reinstated mechanical and heat hypersensitivity. We first tested the hypothesis that LS extends to serotonergic neurons in the rostral ventral medulla (RVM) that convey pronociceptive input to the spinal cord. We report that in male and female mice, hypersensitivity was accompanied by increased Fos expression in serotonergic neurons of the RVM, abolished on chemogenetic inhibition of RVM 5-HT neurons, and blocked by intrathecal injection of the 5-HT3R antagonist ondansetron; the 5-HT2AR antagonist MDL-11 939 had no effect. Second, to test for MORCA, we microinjected the MOR inverse agonist d-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP) and/or neutral opioid receptor antagonist 6β-naltrexol. Intra-RVM CTAP produced mechanical hypersensitivity at both hindpaws; 6β-naltrexol had no effect by itself, but blocked CTAP-induced hypersensitivity. This indicates that MORCA, rather than an opioid ligand-dependent mechanism, maintains LS in remission. We conclude that incision establishes LS in descending RVM 5-HT neurons that drives pronociceptive 5-HT3R signaling in the dorsal horn, and this LS is tonically opposed by MORCA in the RVM. The 5-HT3 receptor is a promising therapeutic target for the development of drugs to prevent the transition from acute to chronic postsurgical pain.SIGNIFICANCE STATEMENT Surgery leads to latent pain sensitization and a compensatory state of endogenous pain control that is maintained long after tissue healing. Here, we show that either chemogenetic inhibition of serotonergic neuron activity in the RVM or pharmacological inhibition of 5-HT3 receptor signaling at the spinal cord blocks behavioral signs of postsurgical latent sensitization. We conclude that MORCA in the RVM opposes descending serotonergic facilitation of LS and that the 5-HT3 receptor is a promising therapeutic target for the development of drugs to prevent the transition from acute to chronic postsurgical pain.
Collapse
Affiliation(s)
- Andrew H Cooper
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Naomi S Hedden
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Pranav Prasoon
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Yanmei Qi
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Bradley K Taylor
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
9
|
Mo 莫思怡 SY, Xu 徐啸翔 XX, Bai 白珊珊 SS, Liu 刘云 Y, Fu 傅开元 KY, Sessle BJ, Cao 曹烨 Y, Xie 谢秋菲 QF. Neuronal Activities in the Rostral Ventromedial Medulla Associated with Experimental Occlusal Interference-Induced Orofacial Hyperalgesia. J Neurosci 2022; 42:5314-5329. [PMID: 35667852 PMCID: PMC9270923 DOI: 10.1523/jneurosci.0008-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/14/2022] [Accepted: 05/24/2022] [Indexed: 01/09/2023] Open
Abstract
The imbalanced conditions of pronociceptive ON-cells and antinociceptive OFF-cells in the rostral ventromedial medulla (RVM) alter nociceptive transmission and play an important role in the development of chronic pain. This study aimed to explore the neuroplastic mechanisms of the RVM ON-cells and OFF-cells in a male rat model of experimental occlusal interference (EOI)-induced nociceptive behavior reflecting orofacial hyperalgesia and in modified models involving EOI removal at early and later stages. We recorded the mechanical head withdrawal thresholds, orofacial operant behaviors, and the activity of identified RVM ON-cells and OFF-cells in these rats. EOI-induced orofacial hyperalgesia could be relieved by EOI removal around postoperative day 3; this effect could be inhibited by intra-RVM microinjection of the κ-opioid receptor agonist U-69593. EOI removal around postoperative day 8 did not relieve the orofacial hyperalgesia, which could, however, be reversed by intra-RVM microinjection of the NK-1 (neurokinin-1) receptor antagonist L-733060. The activity of ON-cells and OFF-cells did not change during both the initial 3 and 6 d of EOI. When EOI was removed on postoperative day 3, OFF-cell responses decreased, contributing to the reversal of hyperalgesia. When EOI lasted for 8 d or was removed on postoperative day 8, spontaneous activity and stimulus-evoked responses of ON-cell increased, contributing to the maintained hyperalgesia. In contrast, when the EOI lasted for 14 d, OFF-cell responses decreased, possibly participating in the maintenance of hyperalgesia with persistent EOI. Our results reveal that adaptive changes in the RVM were associated with orofacial pain following EOI placement and removal.SIGNIFICANCE STATEMENT A considerable proportion of patients experience chronic orofacial pain throughout life despite the therapies given or removal of potential etiologic factors. However, current therapies lack effectiveness because of limited knowledge of the chronicity mechanisms. Using electrophysiological recording, combined with a behavioral test, we found that the prevailing descending facilitation in the rostral ventromedial medulla (RVM) participates in the maintenance of orofacial hyperalgesia following late removal of nociceptive stimuli, while the prevailing descending inhibition from the RVM may contribute to the reversal of orofacial hyperalgesia following early removal of nociceptive stimuli. Thus, variable clinical outcomes of orofacial pain may be associated with descending modulation, and an optimal window of time may exist in the management of chronic orofacial pain.
Collapse
Affiliation(s)
- Si-Yi Mo 莫思怡
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
| | - Xiao-Xiang Xu 徐啸翔
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
| | - Shan-Shan Bai 白珊珊
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
| | - Yun Liu 刘云
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
| | - Kai-Yuan Fu 傅开元
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
- Center for Temporomandibular Disorders and Orofacial Pain, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
| | - Barry J Sessle
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Centre for the Study of Pain, University of Toronto, Toronto, Ontario M5T 1P8, Canada
| | - Ye Cao 曹烨
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
| | - Qiu-Fei Xie 谢秋菲
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, School and Hospital of Stomatology, Peking University, Beijing 100081, People's Republic of China
- National Engineering Research Center for Oral Biomaterials and Digital Medical Devices, Beijing 100081, People's Republic of China
- National Clinical Research Center for Oral Diseases, Beijing 100081, People's Republic of China
| |
Collapse
|
10
|
Patel R, Lumb BM, Bannister K. Editorial: Plasticity of Endogenous Pain Modulatory Circuits in Neuropathy. FRONTIERS IN PAIN RESEARCH 2022; 2:776948. [PMID: 35295507 PMCID: PMC8915642 DOI: 10.3389/fpain.2021.776948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 12/03/2022] Open
Affiliation(s)
- Ryan Patel
- Wolfson Centre for Age Related Diseases, King's College London, London, United Kingdom
| | - Bridget M Lumb
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Kirsty Bannister
- Wolfson Centre for Age Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
11
|
Single-Dose P2 X4R Single-Chain Fragment Variable Antibody Permanently Reverses Chronic Pain in Male Mice. Int J Mol Sci 2021; 22:ijms222413612. [PMID: 34948407 PMCID: PMC8706307 DOI: 10.3390/ijms222413612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022] Open
Abstract
Non-opioid single-chain variable fragment (scFv) small antibodies were generated as pain-reducing block of P2X4R receptor (P2X4R). A panel of scFvs targeting an extracellular peptide sequence of P2X4R was generated followed by cell-free ribosome display for recombinant antibody selection. After three rounds of bio-panning, a panel of recombinant antibodies was isolated and characterized by ELISA, cross-reactivity analysis, and immunoblotting/immunostaining. Generated scFv antibodies feature binding activity similar to monoclonal antibodies but with stronger affinity and increased tissue penetrability due to their ~30% smaller size. Two anti-P2X4R scFv clones (95, 12) with high specificity and affinity binding were selected for in vivo testing in male and female mice with trigeminal nerve chronic neuropathic pain (FRICT-ION model) persisting for several months in untreated BALBc mice. A single dose of P2X4R scFv (4 mg/kg, i.p.) successfully, completely, and permanently reversed chronic neuropathic pain-like measures in male mice only, providing retention of baseline behaviors indefinitely. Untreated mice retained hypersensitivity, and developed anxiety- and depression-like behaviors within 5 weeks. In vitro P2X4R scFv 95 treatment significantly increased the rheobase of larger-diameter (>25 µm) trigeminal ganglia (TG) neurons from FRICT-ION mice compared to controls. The data support use of engineered scFv antibodies as non-opioid biotherapeutic interventions for chronic pain.
Collapse
|
12
|
Puja G, Sonkodi B, Bardoni R. Mechanisms of Peripheral and Central Pain Sensitization: Focus on Ocular Pain. Front Pharmacol 2021; 12:764396. [PMID: 34916942 PMCID: PMC8669969 DOI: 10.3389/fphar.2021.764396] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
Persistent ocular pain caused by corneal inflammation and/or nerve injury is accompanied by significant alterations along the pain axis. Both primary sensory neurons in the trigeminal nerves and secondary neurons in the spinal trigeminal nucleus are subjected to profound morphological and functional changes, leading to peripheral and central pain sensitization. Several studies using animal models of inflammatory and neuropathic ocular pain have provided insight about the mechanisms involved in these maladaptive changes. Recently, the advent of new techniques such as optogenetics or genetic neuronal labelling has allowed the investigation of identified circuits involved in nociception, both at the spinal and trigeminal level. In this review, we will describe some of the mechanisms that contribute to the perception of ocular pain at the periphery and at the spinal trigeminal nucleus. Recent advances in the discovery of molecular and cellular mechanisms contributing to peripheral and central pain sensitization of the trigeminal pathways will be also presented.
Collapse
Affiliation(s)
- Giulia Puja
- Department of Life Sciences, University of Modena and Reggio Emilia, Emilia-Romagna, Italy
| | - Balazs Sonkodi
- Department of Health Sciences and Sport Medicine, University of Physical Education, Budapest, Hungary
| | - Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Emilia-Romagna, Italy
| |
Collapse
|
13
|
Zheng H, Lim JY, Kim Y, Jung ST, Hwang SW. The role of oxytocin, vasopressin, and their receptors at nociceptors in peripheral pain modulation. Front Neuroendocrinol 2021; 63:100942. [PMID: 34437871 DOI: 10.1016/j.yfrne.2021.100942] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/01/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Oxytocin and vasopressin are neurohypophyseal hormones with sequence similarity and play a central role in bodily homeostatic regulation. Pain is currently understood to be an important phenotype that those two neurohormones strongly downregulate. Nociceptors, the first component of the ascending neural circuit for pain signals, have constantly been shown to be modulated by those peptides. The nociceptor modulation appears to be critical in pain attenuation, which has led to a gradual increase in scientific interest about their physiological processes and also drawn attention to their translational potentials. This review focused on what are recently understood and stay under investigation in the functional modulation of nociceptors by oxytocin and vasopressin. Effort to produce a nociceptor-specific view could help to construct a more systematic picture of the peripheral pain modulation by oxytocin and vasopressin.
Collapse
Affiliation(s)
- Haiyan Zheng
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; Department of Physiology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Ji Yeon Lim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; Department of Physiology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Yerin Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; Department of Physiology, College of Medicine, Korea University, Seoul 02841, Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Sun Wook Hwang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; Department of Physiology, College of Medicine, Korea University, Seoul 02841, Korea.
| |
Collapse
|
14
|
Tavares I, Costa-Pereira JT, Martins I. Monoaminergic and Opioidergic Modulation of Brainstem Circuits: New Insights Into the Clinical Challenges of Pain Treatment? FRONTIERS IN PAIN RESEARCH 2021; 2:696515. [PMID: 35295506 PMCID: PMC8915776 DOI: 10.3389/fpain.2021.696515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022] Open
Abstract
The treatment of neuropathic pain remains a clinical challenge. Analgesic drugs and antidepressants are frequently ineffective, and opioids may induce side effects, including hyperalgesia. Recent results on brainstem pain modulatory circuits may explain those clinical challenges. The dual action of noradrenergic (NA) modulation was demonstrated in animal models of neuropathic pain. Besides the well-established antinociception due to spinal effects, the NA system may induce pronociception by directly acting on brainstem pain modulatory circuits, namely, at the locus coeruleus (LC) and medullary dorsal reticular nucleus (DRt). The serotoninergic system also has a dual action depending on the targeted spinal receptor, with an exacerbated activity of the excitatory 5-hydroxytryptamine 3 (5-HT3) receptors in neuropathic pain models. Opioids are involved in the modulation of descending modulatory circuits. During neuropathic pain, the opioidergic modulation of brainstem pain control areas is altered, with the release of enhanced local opioids along with reduced expression and desensitization of μ-opioid receptors (MOR). In the DRt, the installation of neuropathic pain increases the levels of enkephalins (ENKs) and induces desensitization of MOR, which may enhance descending facilitation (DF) from the DRt and impact the efficacy of exogenous opioids. On the whole, the data discussed in this review indicate the high plasticity of brainstem pain control circuits involving monoaminergic and opioidergic control. The data from studies of these neurochemical systems in neuropathic models indicate the importance of designing drugs that target multiple neurochemical systems, namely, maximizing the antinociceptive effects of antidepressants that inhibit the reuptake of serotonin and noradrenaline and preventing desensitization and tolerance of MOR at the brainstem.
Collapse
Affiliation(s)
- Isaura Tavares
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
- Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
- *Correspondence: Isaura Tavares
| | - José Tiago Costa-Pereira
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
- Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
- Faculty of Nutrition and Food Science, University of Porto, Porto, Portugal
| | - Isabel Martins
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
- Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal
| |
Collapse
|
15
|
Cramer N, Silva-Cardoso G, Masri R, Keller A. Control of synaptic transmission and neuronal excitability in the parabrachial nucleus. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 9:100057. [PMID: 33364528 PMCID: PMC7753201 DOI: 10.1016/j.ynpai.2020.100057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/16/2022]
Abstract
The parabrachial nucleus (PB) is a hub for aversive behaviors, including those related to pain. We have shown that the expression of chronic pain is causally related to amplified activity of PB neurons, and to changes in synaptic inhibition of these neurons. These findings indicate that regulation of synaptic activity in PB may modulate pain perception and be involved in the pathophysiology of chronic pain. Here, we identify the roles in PB of signaling pathways that modulate synaptic functions. In pharmacologically isolated lateral PB neurons in acute mouse slices we find that baclofen, a GABAB receptor agonist, suppresses the frequency of miniature inhibitory and excitatory postsynaptic currents (mIPSCs and mEPSC). Activation of µ-opioid peptide receptors with DAMGO had similar suppressive effects on excitatory and inhibitory synapses, while the κ-opioid peptide receptor agonist U-69593 suppressed mIPSC release but had no consistent effects on mEPSCs. Activation of cannabinoid type 1 receptors with WIN 55,212-2 reduced the frequency of both inhibitory and excitatory synaptic events, while the CB1 receptor inverse agonist AM251 had opposite effects on mIPSC and mEPSC frequencies. AM251 increased the frequency of inhibitory events but led to a reduction in excitatory events through a GABAB mediated mechanism. Although none of the treatments produced a consistent effect on mIPSC or mEPSC amplitudes, baclofen and DAMGO both reliably activated a postsynaptic conductance. These results demonstrate that multiple signaling pathways can alter synaptic transmission and neuronal excitability in PB and provide a basis for investigating the contributions of these systems to the development and maintenance of chronic pain.
Collapse
Affiliation(s)
- Nathan Cramer
- Department of Anatomy and Neurobiology and the Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Gleice Silva-Cardoso
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Brazil
| | - Radi Masri
- Department of Oral Sciences and Pain. School of Dentistry, University of Maryland Baltimore, Baltimore, MD, USA
| | - Asaf Keller
- Department of Anatomy and Neurobiology and the Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Ren K. Grand Challenges in Musculoskeletal Pain Research: Chronicity, Comorbidity, Immune Regulation, Sex Differences, Diagnosis, and Treatment Opportunities. FRONTIERS IN PAIN RESEARCH 2020; 1. [PMID: 34296207 PMCID: PMC8294784 DOI: 10.3389/fpain.2020.575479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, United States.,Program in Neuroscience, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
17
|
Li JH, Yang JL, Wei SQ, Li ZL, Collins AA, Zou M, Wei F, Cao DY. Contribution of central sensitization to stress-induced spreading hyperalgesia in rats with orofacial inflammation. Mol Brain 2020; 13:106. [PMID: 32723345 PMCID: PMC7385893 DOI: 10.1186/s13041-020-00645-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Temporomandibular disorder (TMD) is commonly comorbid with fibromyalgia syndrome (FMS). The incidence of these pain conditions is prevalent in women and prone to mental stress. Chronic pain symptoms in patients with FMS and myofascial TMD (mTMD) are severe and debilitating. In the present study, we developed a new animal model to mimic the comorbidity of TMD and FMS. In ovariectomized female rats, repeated forced swim (FS) stress induced mechanical allodynia and thermal hyperalgesia in the hindpaws of the 17β-estradiol (E2) treated rats with orofacial inflammation. Subcutaneous injection of E2, injection of complete Freund’s adjuvant (CFA) into masseter muscles or FS alone did not induce somatic hyperalgesia. We also found that the somatic hyperalgesia was accompanied by upregulation of GluN1 receptor and serotonin (5-hydroxytryptamine, 5-HT)3A receptor expression in the dorsal horn of spinal cord at L4-L5 segments. Intrathecal injection of N-methyl-D-aspartic acid receptor (NMDAR) antagonist 2-amino-5-phosphonovaleric acid (APV) or 5-HT3 receptor antagonist Y-25130 blocked stress-induced wide-spreading hyperalgesia. These results suggest that NMDAR-dependent central sensitization in the spinal dorsal horn and 5-HT-dependent descending facilitation contribute to the development of wide-spreading hyperalgesia in this comorbid pain model.
Collapse
Affiliation(s)
- Jia-Heng Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China.,Department of Orthodontics, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China.,Department of Neural and Pain Sciences, University of Maryland School of Dentistry; the UM Center to Advance Chronic Pain Research, 650 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Jia-Le Yang
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry; the UM Center to Advance Chronic Pain Research, 650 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Si-Qi Wei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Zhuo-Lin Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Anna A Collins
- Department of Orthodontics, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Min Zou
- Department of Orthodontics, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Feng Wei
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry; the UM Center to Advance Chronic Pain Research, 650 West Baltimore Street, Baltimore, MD, 21201, USA.
| | - Dong-Yuan Cao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China.
| |
Collapse
|
18
|
Lee GW, Son JY, Lee AR, Ju JS, Bae YC, Ahn DK. Central VEGF-A pathway plays a key role in the development of trigeminal neuropathic pain in rats. Mol Pain 2020; 15:1744806919872602. [PMID: 31397622 PMCID: PMC6719481 DOI: 10.1177/1744806919872602] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The study reported here investigated the role of the central vascular endothelial growth factor-A (VEGF-A) pathway in the development of trigeminal neuropathic pain following nerve injury. A Sprague-Dawley rat model of trigeminal neuropathic pain was produced using malpositioned dental implants. The left mandibular second molar was extracted under anesthesia and replaced with a miniature dental implant to induce injury to the inferior alveolar nerve. The inferior alveolar nerve injury produced a significant upregulation of astrocytic VEGF-A expression in the medullary dorsal horn. The nerve injury-induced mechanical allodynia was inhibited by an intracisternal infusion of VEGF-A164 antibody. Although both VEGF-A Receptor 1 (VEGF-A R1; colocalized with the blood–brain barrier) and VEGF-A Receptor 2 (VEGF-A R2; colocalized with astrocytes) participated in the development of trigeminal neuropathic pain following nerve injury, only the intracisternal infusion of a VEGF-A R1 antibody, and not that of a VEGF-A R2 antibody, inhibited the increased blood–brain barrier permeability produced by nerve injury. Finally, we confirmed the participation of the central VEGF-A pathway in the development of trigeminal neuropathic pain by reducing VEGF-A expression using VEGF-A164 siRNA. This suppression of VEGF-A produced significant prolonged anti-allodynic effects. These results suggest that the central VEGF-A pathway plays a key role in the development of trigeminal neuropathic pain following nerve injury through two separate pathways: VEGF-A R1 and VEGF-A R2. Hence, a blockade of the central VEGF-A pathway provides a new therapeutic avenue for the treatment of trigeminal neuropathic pain.
Collapse
Affiliation(s)
- Geun W Lee
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jo Y Son
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Ah R Lee
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jin S Ju
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Yong C Bae
- 2 Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Dong K Ahn
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| |
Collapse
|
19
|
Ablation of TRPV1+ Afferent Terminals by Capsaicin Mediates Long-Lasting Analgesia for Trigeminal Neuropathic Pain. eNeuro 2020; 7:ENEURO.0118-20.2020. [PMID: 32404326 PMCID: PMC7266139 DOI: 10.1523/eneuro.0118-20.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
Trigeminal neuropathic pain (TNP) is often resistant to current pharmacotherapy, and there is a pressing need to develop more efficacious treatments. Capsaicin is a pungent ingredient of chili peppers and specifically activates transient receptor potential vanilloid subtype 1 (TRPV1), a Ca2+-permeable ion channel. Topical capsaicin invariably induces burning pain. Paradoxically, the transient pain is often followed by prolonged attenuation of the preexisting pathologic pain from the same region. However, the mechanisms underlying capsaicin-induced analgesia are not well understood. Although the reports of the involvement of TRPV1 and TRPV1+ afferents in neuropathic pain are controversial, we recently demonstrated that TRPV1 and TRPV1+ afferents are involved in mechanical hyperalgesia in mice with chronic constriction injury of the infraorbital nerve (ION-CCI). Consistently, chemogenetic inhibition of TRPV1-lineage (TRPV1-LN) afferents attenuated mechanical hyperalgesia and ongoing pain. In mice with ION-CCI, we found that a single focal injection of capsaicin into facial skin led to attenuation of mechanical hyperalgesia over two weeks. Capsaicin treatment also attenuated secondary hyperalgesia in extraterritorial mandibular skin. Furthermore, capsaicin treatment decreased ongoing pain. Longitudinal in vivo two-photon imaging of cutaneous nerve fibers showed that such capsaicin-induced analgesia is correlated with cutaneous nerve terminal density. Furthermore, preventing capsaicin-induced ablation of afferent terminals by co-administration of capsaicin with MDL28170, an inhibitor of calpain, abolished capsaicin-induced analgesia. These results suggest that a single focal injection of capsaicin induces long-lasting analgesia for neuropathic pain via selective ablation of TRPV1+ afferent terminals and that TRPV1+ afferents contribute to the maintenance of TNP.
Collapse
|
20
|
Chung MK, Wang S, Yang J, Alshanqiti I, Wei F, Ro JY. Neural Pathways of Craniofacial Muscle Pain: Implications for Novel Treatments. J Dent Res 2020; 99:1004-1012. [PMID: 32374638 DOI: 10.1177/0022034520919384] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Craniofacial muscle pain is highly prevalent in temporomandibular disorders but is difficult to treat. Enhanced understanding of neurobiology unique to craniofacial muscle pain should lead to the development of novel mechanism-based treatments. Herein, we review recent studies to summarize neural pathways of craniofacial muscle pain. Nociceptive afferents in craniofacial muscles are predominantly peptidergic afferents enriched with TRPV1. Signals from peripheral glutamate receptors converge onto TRPV1, leading to mechanical hyperalgesia. Further studies are needed to clarify whether hyperalgesic priming in nonpeptidergic afferents or repeated acid injections also affect craniofacial muscle pain. Within trigeminal ganglia, afferents innervating craniofacial muscles interact with surrounding satellite glia, which enhances the sensitivity of the inflamed neurons as well as nearby uninjured afferents, resulting in hyperalgesia and ectopic pain originating from adjacent orofacial tissues. Craniofacial muscle afferents project to a wide area within the trigeminal nucleus complex, and central sensitization of medullary dorsal horn neurons is a critical factor in muscle hyperalgesia related to ectopic pain and emotional stress. Second-order neurons project rostrally to pathways associated with affective pain, such as parabrachial nucleus and medial thalamic nucleus, as well as sensory-discriminative pain, such as ventral posteromedial thalamic nuclei. Abnormal endogenous pain modulation can also contribute to chronic muscle pain. Descending serotonergic circuits from the rostral ventromedial medulla facilitate activation of second-order neurons in the trigeminal nucleus complex, which leads to the maintenance of mechanical hyperalgesia of inflamed masseter muscle. Patients with temporomandibular disorders exhibit altered brain networks in widespread cortical and subcortical regions. Recent development of methods for neural circuit manipulation allows silencing of specific hyperactive neural circuits. Chemogenetic silencing of TRPV1-expressing afferents or rostral ventromedial medulla neurons attenuates hyperalgesia during masseter inflammation. It is likely, therefore, that further delineation of neural circuits mediating craniofacial muscle hyperalgesia potentially enhances treatment of chronic muscle pain conditions.
Collapse
Affiliation(s)
- M K Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, USA
| | - S Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, USA
| | - J Yang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, USA
| | - I Alshanqiti
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, USA
| | - F Wei
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, USA
| | - J Y Ro
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, USA
| |
Collapse
|
21
|
Raver C, Uddin O, Ji Y, Li Y, Cramer N, Jenne C, Morales M, Masri R, Keller A. An Amygdalo-Parabrachial Pathway Regulates Pain Perception and Chronic Pain. J Neurosci 2020; 40:3424-3442. [PMID: 32217613 PMCID: PMC7178908 DOI: 10.1523/jneurosci.0075-20.2020] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
The parabrachial (PB) complex mediates both ascending nociceptive signaling and descending pain modulatory information in the affective/emotional pain pathway. We have recently reported that chronic pain is associated with amplified activity of PB neurons in a rat model of neuropathic pain. Here we demonstrate that similar activity amplification occurs in mice, and that this is related to suppressed inhibition to lateral parabrachial (LPB) neurons from the CeA in animals of either sex. Animals with pain after chronic constriction injury of the infraorbital nerve (CCI-Pain) displayed higher spontaneous and evoked activity in PB neurons, and a dramatic increase in after-discharges, responses that far outlast the stimulus, compared with controls. LPB neurons in CCI-Pain animals showed a reduction in inhibitory, GABAergic inputs. We show that, in both rats and mice, LPB contains few GABAergic neurons, and that most of its GABAergic inputs arise from CeA. These CeA GABA neurons express dynorphin, somatostatin, and/or corticotropin releasing hormone. We find that the efficacy of this CeA-LPB pathway is suppressed in chronic pain. Further, optogenetically stimulating this pathway suppresses acute pain, and inhibiting it, in naive animals, evokes pain behaviors. These findings demonstrate that the CeA-LPB pathway is critically involved in pain regulation, and in the pathogenesis of chronic pain.SIGNIFICANCE STATEMENT We describe a novel pathway, consisting of inhibition by dynorphin, somatostatin, and corticotropin-releasing hormone-expressing neurons in the CeA that project to the parabrachial nucleus. We show that this pathway regulates the activity of pain-related neurons in parabrachial nucleus, and that, in chronic pain, this inhibitory pathway is suppressed, and that this suppression is causally related to pain perception. We propose that this amygdalo-parabrachial pathway is a key regulator of both chronic and acute pain, and a novel target for pain relief.
Collapse
Affiliation(s)
- Charles Raver
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Olivia Uddin
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yadong Ji
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, Maryland 21201
| | - Ying Li
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Nathan Cramer
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Carleigh Jenne
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Marisela Morales
- Neuronal Networks Section, Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland 21224
| | - Radi Masri
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, Maryland 21201
| | - Asaf Keller
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
22
|
Li M, Zhu M, Xu Q, Ding F, Tian Y, Zhang M. Sensation of TRPV1 via 5-hydroxytryptamine signaling modulates pain hypersensitivity in a 6-hydroxydopamine induced mice model of Parkinson’s disease. Biochem Biophys Res Commun 2020; 521:868-873. [DOI: 10.1016/j.bbrc.2019.10.204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022]
|
23
|
Calcium Channel α2δ1 Subunit Mediates Secondary Orofacial Hyperalgesia Through PKC-TRPA1/Gap Junction Signaling. THE JOURNAL OF PAIN 2020; 21:238-257. [DOI: 10.1016/j.jpain.2019.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/06/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
|
24
|
Shinoda M, Kubo A, Hayashi Y, Iwata K. Peripheral and Central Mechanisms of Persistent Orofacial Pain. Front Neurosci 2019; 13:1227. [PMID: 31798407 PMCID: PMC6863776 DOI: 10.3389/fnins.2019.01227] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/30/2019] [Indexed: 12/25/2022] Open
Abstract
Neuroplastic changes in the neuronal networks involving the trigeminal ganglion (TG), trigeminal spinal subnucleus caudalis (Vc), and upper cervical spinal cord (C1/C2) are considered the mechanisms underlying the ectopic orofacial hypersensitivity associated with trigeminal nerve injury or orofacial inflammation. It has been reported that peripheral nerve injury causes injury discharges in the TG neurons, and a barrage of action potentials is generated in TG neurons and conveyed to the Vc and C1/C2 after trigeminal nerve injury. Long after trigeminal nerve injury, various molecules are produced in the TG neurons, and these molecules are released from the soma of TG neurons and are transported to the central and peripheral terminals of TG neurons. These changes within the TG cause neuroplastic changes in TG neurons and they become sensitized. The neuronal activity of TG neurons is further accelerated, and Vc and C1/C2 neurons are also sensitized. In addition to this cascade, non-neuronal glial cells are also involved in the enhancement of the neuronal activity of TG, Vc, and C1/C2 neurons. Satellite glial cells and macrophages are activated in the TG after trigeminal nerve injury and orofacial inflammation. Microglial cells and astrocytes are also activated in the Vc and C1/C2 regions. It is considered that functional interaction between non-neuronal cells and neurons in the TG, Vc, and C1/C2 regions is a key mechanism involved in the enhancement of neuronal excitability after nerve injury or inflammation. In this article, the detailed mechanisms underlying ectopic orofacial hyperalgesia associated with trigeminal nerve injury and orofacial inflammation are addressed.
Collapse
Affiliation(s)
- Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Asako Kubo
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
25
|
Enhanced descending pain facilitation in acute traumatic brain injury. Exp Neurol 2019; 320:112976. [PMID: 31185197 DOI: 10.1016/j.expneurol.2019.112976] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/25/2019] [Accepted: 06/06/2019] [Indexed: 01/23/2023]
Abstract
Acute and persistent pain are recognized consequences of TBI that can enhance suffering and significantly impair rehabilitative efforts. Both experimental models and clinical studies suggest that TBI may result in an imbalance between descending pain facilitatory and inhibitory pathways. The aim of this study was to assess the role of enhanced descending serotonin-mediated pain facilitation in a rat TBI model using selective spinal serotonergic fiber depletion with 5, 7-dihydroxytryptamine (DHT). We observed significant hindpaw allodynia in TBI rats that was reduced after DHT but not vehicle treatment. Immunohistochemical studies demonstrated profound spinal serotonin depletion in DHT-treated rats. Furthermore, lumbar intrathecal administration of the 5-HT3 receptor antagonist ondansetron at 7 days post-injury (DPI), when hindpaw allodynia was maximal, also attenuated nociceptive sensitization. Additional immunohistochemical analyses of the lumbar spinal cord at 7 DPI revealed a robust bilateral microglial response in the superficial dorsal horns that was significantly reduced with DHT treatment. Furthermore, serotonin depletion also prevented the TBI-induced bilateral increase in c-Fos positive cells within the Rexed laminae I and II of the dorsal horns. These results indicate that in the weeks following TBI, pain may be responsive to 5-HT3 receptor antagonists or other measures which rebalance descending pain modulation.
Collapse
|
26
|
Bista P, Imlach WL. Pathological Mechanisms and Therapeutic Targets for Trigeminal Neuropathic Pain. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E91. [PMID: 31443547 PMCID: PMC6789505 DOI: 10.3390/medicines6030091] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/31/2022]
Abstract
Trigeminal neuropathic pain is a chronic pain condition caused by damage or inflammation of the trigeminal nerve or its branches, with both peripheral and central nervous system dysfunction contributing to the disorder. Trigeminal pain conditions present with diagnostic and therapeutic challenges to healthcare providers and often require multiple therapeutic approaches for pain reduction. This review will provide the overview of pathophysiology in peripheral and central nociceptive circuits that are involved in neuropathic pain conditions involving the trigeminal nerve and the current therapeutics that are used to treat these disorders. Recent advances in treatment of trigeminal pain, including novel therapeutics that target ion channels and receptors, gene therapy and monoclonal antibodies that have shown great promise in preclinical studies and clinical trials will also be described.
Collapse
Affiliation(s)
- Pawan Bista
- Department of Physiology & Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Wendy L Imlach
- Department of Physiology & Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
27
|
Akintola T, Tricou C, Raver C, Castro A, Colloca L, Keller A. In search of a rodent model of placebo analgesia in chronic orofacial neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100033. [PMID: 31223137 PMCID: PMC6565753 DOI: 10.1016/j.ynpai.2019.100033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/18/2019] [Accepted: 05/13/2019] [Indexed: 12/25/2022]
Abstract
All treatments are given in a context, suggesting that conditioning cues may significantly influence therapeutic outcomes. We tested the hypothesis that context affects placebo analgesia in rodents. To produce neuropathic pain in rats, we performed chronic constriction injury of the infraorbital nerve. We then treated the rats daily, over a seven day period, with injections of either fentanyl or saline, with or without associated conditioning cues; a fourth group received no treatment. On the eighth day, we replaced fentanyl with saline to test for conditioned placebo analgesia. We tested the effects of treatment by measuring sensitivity to mechanical stimuli and grimace scale scores. We found no significant differences in either of these outcomes among the four experimental groups. These findings suggest that chronic, neuropathic pain in rats may not be susceptible to placebo analgesia.
Collapse
Affiliation(s)
- Titilola Akintola
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Program in Toxicology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, USA
| | - Christina Tricou
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Charles Raver
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alberto Castro
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luana Colloca
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
- Departments of Anesthesiology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Asaf Keller
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Uddin O, Studlack PE, Parihar S, Keledjian K, Cruz A, Farooq T, Shin N, Gerzanich V, Simard JM, Keller A. Chronic pain after blast-induced traumatic brain injury in awake rats. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100030. [PMID: 31223145 PMCID: PMC6565615 DOI: 10.1016/j.ynpai.2019.100030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022]
Abstract
Explosive blast-induced traumatic brain injury (blast-TBI) in military personnel is a leading cause of injury and persistent neurological abnormalities, including chronic pain. We previously demonstrated that chronic pain after spinal cord injury results from central sensitization in the posterior thalamus (PO). The presence of persistent headaches and back pain in veterans with blast-TBI suggests a similar involvement of thalamic sensitization. Here, we tested the hypothesis that pain after blast-TBI is associated with abnormal increases in activity of neurons in PO thalamus. We developed a novel model with two unique features: (1) blast-TBI was performed in awake, un-anesthetized rats, to simulate the human experience and to eliminate confounds of anesthesia and surgery inherent in other models; (2) only the cranium, rather than the entire body, was exposed to a collimated blast wave, with the blast wave striking the posterior cranium in the region of the occipital crest and foramen magnum. Three weeks after blast-TBI, rats developed persistent, ongoing spontaneous pain. Contrary to our hypothesis, we found no significant differences in the activity of PO neurons, or of neurons in the spinal trigeminal nucleus. There were also no significant changes in gliosis in either of these structures. This novel model will allow future studies on the pathophysiology of chronic pain after blast-TBI.
Collapse
Affiliation(s)
- Olivia Uddin
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Paige E. Studlack
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Saitu Parihar
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
| | - Alexis Cruz
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Tayyiaba Farooq
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Naomi Shin
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
- Department of Pathology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD, USA
| | - Asaf Keller
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| |
Collapse
|
29
|
Liu S, Tang Y, Shu H, Tatum D, Bai Q, Crawford J, Xing Y, Lobo MK, Bellinger L, Kramer P, Tao F. Dopamine receptor D2, but not D1, mediates descending dopaminergic pathway-produced analgesic effect in a trigeminal neuropathic pain mouse model. Pain 2019; 160:334-344. [PMID: 30325872 PMCID: PMC6344251 DOI: 10.1097/j.pain.0000000000001414] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neuropathic pain represents a challenge to clinicians because it is resistant to commonly prescribed analgesics due to its largely unknown mechanisms. Here, we investigated a descending dopaminergic pathway-mediated modulation of trigeminal neuropathic pain. We performed chronic constriction injury of the infraorbital nerve from the maxillary branch of trigeminal nerve to induce trigeminal neuropathic pain in mice. Our retrograde tracing showed that the descending dopaminergic projection from hypothalamic A11 nucleus to spinal trigeminal nucleus caudalis is bilateral. Optogenetic/chemogenetic manipulation of dopamine receptors D1 and D2 in the spinal trigeminal nucleus caudalis produced opposite effects on the nerve injury-induced trigeminal neuropathic pain. Specific excitation of dopaminergic neurons in the A11 nucleus attenuated the trigeminal neuropathic pain through the activation of D2 receptors in the spinal trigeminal nucleus caudalis. Conversely, specific ablation of the A11 dopaminergic neurons exacerbated such pain. Our results suggest that the descending A11-spinal trigeminal nucleus caudalis dopaminergic projection is critical for the modulation of trigeminal neuropathic pain and could be manipulated to treat such pain.
Collapse
Affiliation(s)
- Sufang Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China
| | - Yuanyuan Tang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hui Shu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Delton Tatum
- Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Qian Bai
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Joshua Crawford
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Ying Xing
- Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou, Henan, China
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Larry Bellinger
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Phillip Kramer
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
- Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, Texas, USA
| |
Collapse
|
30
|
Guo W, Imai S, Zou S, Yang J, Watanabe M, Wang J, Dubner R, Wei F, Ren K. Altered glial glutamate transporter expression in descending circuitry and the emergence of pain chronicity. Mol Pain 2019; 15:1744806918825044. [PMID: 30799685 PMCID: PMC6348548 DOI: 10.1177/1744806918825044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The glutamate type 1 transporter (GLT1) plays a major role in glutamate homeostasis in the brain. Although alterations of GLT1 activity have been linked to persistent pain, the significance of these changes is poorly understood. Focusing on the rostral ventromedial medulla, a key site in pain modulation, we examined the expression and function of GLT1 and related transcription factor kappa B-motif binding phosphoprotein (KBBP) in rats after adjuvant-induced hind paw inflammation. RESULTS After inflammation, GLT1 and KBBP showed an early upregulation and gradual transition to downregulation that lasted throughout the eight-week observation period. Nitration of GLT1 was reduced at 30 min and increased at eight weeks after inflammation, suggesting an initial increase and later decrease in transporter activity. Mechanical hyperalgesia and paw edema exhibited an initial developing phase with peak hyperalgesia at 4 to 24 h, a subsequent attenuating phase, followed by a late persistent phase that lasted for months. The downregulation of GLT1 occurred at a time when hyperalgesia transitioned into the persistent phase. In the rostral ventromedial medulla, pharmacological block with dihydrokainic acid and RNAi of GLT1 and KBBP increased nociception and overexpression of GLT1 reversed persistent hyperalgesia. Further, the initial upregulation of GLT1 and KBBP was blocked by local anesthetic block, and pretreatment with dihydrokainic acid facilitated the development of hyperalgesia. CONCLUSIONS These results suggest that the initial increased GLT1 activity depends on injury input and serves to dampen the development of hyperalgesia. However, later downregulation of GLT1 fosters the net descending facilitation as injury persists, leading to the emergence of persistent pain.
Collapse
Affiliation(s)
- Wei Guo
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Satoshi Imai
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Shiping Zou
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Jiale Yang
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Mineo Watanabe
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
- 3 Department of Oral Biology, Division of Molecular Medical Science, Hiroshima, Japan
| | - Jing Wang
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
- 4 Key Laboratory of Bone and Joint Diseases of Gansu province, Institute of Orthopedics, the Second Hospital of Lanzhou University, Lanzhou, China
| | - Ronald Dubner
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Feng Wei
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| | - Ke Ren
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- 2 Program in Neuroscience, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
31
|
Patel R, Dickenson AH. Modality selective roles of pro-nociceptive spinal 5-HT 2A and 5-HT 3 receptors in normal and neuropathic states. Neuropharmacology 2018; 143:29-37. [PMID: 30240783 PMCID: PMC6277848 DOI: 10.1016/j.neuropharm.2018.09.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/23/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022]
Abstract
Descending brainstem control of spinal nociceptive processing permits a dynamic and adaptive modulation of ascending sensory information. Chronic pain states are frequently associated with enhanced descending excitatory drive mediated predominantly through serotonergic neurones in the rostral ventromedial medulla. In this study, we examine the roles of spinal 5-HT2A and 5-HT3 receptors in modulating ascending sensory output in normal and neuropathic states. In vivo electrophysiology was performed in anaesthetised spinal nerve ligated (SNL) and sham-operated rats to record from wide dynamic range neurones in the ventral posterolateral thalamus. In sham rats, block of spinal 5-HT3Rs with ondansetron revealed tonic facilitation of noxious punctate mechanical stimulation, whereas blocking 5-HT2ARs with ketanserin had minimal effect on neuronal responses to evoked stimuli. The inhibitory profiles of both drugs were altered in SNL rats; ondansetron additionally inhibited neuronal responses to lower intensity punctate mechanical stimuli and noxious heat evoked responses, whereas ketanserin inhibited innocuous and noxious evaporative cooling evoked responses. Neither drug had any effect on dynamic brush evoked responses nor on spontaneous firing rates in both sham and SNL rats. These data identify novel modality and intensity selective facilitatory roles of spinal 5-HT2A and 5-HT3 receptors on sensory neuronal processing within the spinothalamic-somatosensory cortical pathway.
Collapse
Affiliation(s)
- Ryan Patel
- University College London, Gower Street, Department of Neuroscience, Physiology and Pharmacology, London, WC1E 6BT, UK.
| | - Anthony H Dickenson
- University College London, Gower Street, Department of Neuroscience, Physiology and Pharmacology, London, WC1E 6BT, UK
| |
Collapse
|
32
|
Studlack PE, Keledjian K, Farooq T, Akintola T, Gerzanich V, Simard JM, Keller A. Blast-induced brain injury in rats leads to transient vestibulomotor deficits and persistent orofacial pain. Brain Inj 2018; 32:1866-1878. [PMID: 30346868 PMCID: PMC6381394 DOI: 10.1080/02699052.2018.1536282] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 09/18/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022]
Abstract
Blast-induced traumatic brain injury (blast-TBI) is associated with vestibulomotor dysfunction, persistent post-traumatic headaches and post-traumatic stress disorder, requiring extensive treatments and reducing quality-of-life. Treatment and prevention of these devastating outcomes require an understanding of their underlying pathophysiology through studies that take advantage of animal models. Here, we report that cranium-directed blast-TBI in rats results in signs of pain that last at least 8 weeks after injury. These occur without significantly elevated behavioural markers of anxiety-like conditions and are not associated with glial up-regulation in sensory thalamic nuclei. These injuries also produce transient vestibulomotor abnormalities that resolve within 3 weeks of injury. Thus, blast-TBI in rats recapitulates aspects of the human condition.
Collapse
Affiliation(s)
- Paige E. Studlack
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St., HSFII S251, Baltimore, MD 21201, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S. Pine St., MSTF 634B, Baltimore, MD 21201, USA
| | - Tayyiaba Farooq
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St., HSFII S251, Baltimore, MD 21201, USA
| | - Titilola Akintola
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St., HSFII S251, Baltimore, MD 21201, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S. Pine St., MSTF 634B, Baltimore, MD 21201, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S. Pine St., MSTF 634B, Baltimore, MD 21201, USA
| | - Asaf Keller
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St., HSFII S251, Baltimore, MD 21201, USA
| |
Collapse
|
33
|
Araújo-Filho HG, Pereira EWM, Campos AR, Quintans-Júnior LJ, Quintans JSS. Chronic orofacial pain animal models - progress and challenges. Expert Opin Drug Discov 2018; 13:949-964. [PMID: 30220225 DOI: 10.1080/17460441.2018.1524458] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Chronic orofacial pain is one of the most common pain conditions experienced by adults. Animal models are often selected as the most useful scientific methodology to explore the pathophysiology of the disorders that cause this disabling pain to facilitate the development of new treatments. The creation of new models or the improvement of existing ones is essential for finding new ways to approach the complex neurobiology of this type of pain. Areas covered: The authors describe and discuss a variety of animal models used in chronic orofacial pain (COFP). Furthermore, they examine in detail the mechanisms of action involved in orofacial neuropathic pain and orofacial inflammatory pain. Expert opinion: The use of animal models has several advantages in chronic orofacial pain drug discovery. Choosing an animal model that most closely represents the human disease helps to increase the chances of finding effective new therapies and is key to the successful translation of preclinical research to clinical practice. Models using genetically modified animals seem promising but have not yet been fully developed for use in chronic orofacial pain research. Although animal models have provided significant advances in the pharmacological treatment of orofacial pain, several barriers still need to be overcome for better treatment options.
Collapse
Affiliation(s)
- Heitor G Araújo-Filho
- a Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology , Federal University of Sergipe , São Cristóvão , Brazil
| | - Erik W M Pereira
- a Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology , Federal University of Sergipe , São Cristóvão , Brazil
| | - Adriana Rolim Campos
- b Experimental Biology Centre (NUBEX) , University of Fortaleza (UNIFOR) , Fortaleza , Brazil
| | - Lucindo J Quintans-Júnior
- a Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology , Federal University of Sergipe , São Cristóvão , Brazil
| | - Jullyana S S Quintans
- a Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology , Federal University of Sergipe , São Cristóvão , Brazil
| |
Collapse
|
34
|
Uddin O, Studlack P, Akintola T, Raver C, Castro A, Masri R, Keller A. Amplified parabrachial nucleus activity in a rat model of trigeminal neuropathic pain. NEUROBIOLOGY OF PAIN 2018; 3:22-30. [PMID: 29862375 PMCID: PMC5973803 DOI: 10.1016/j.ynpai.2018.02.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The parabrachial (PB) complex mediates both ascending nociceptive signaling and descending pain modulatory information in the affective/emotional pain pathway. We hypothesized that PB hyperactivity influences chronic pain behavior after trigeminal nerve injury in rats. Following induction of neuropathic pain using the chronic constriction injury of the infraorbital nerve (CCI-ION) model, rats displayed spontaneous markers of pain and mechanical hyperalgesia extending beyond the receptive field of the injured nerve. PB neurons recorded from rats with CCI-ION displayed amplified activity, manifesting as significantly longer responses to sensory stimuli, compared to shams. These findings suggest that chronic neuropathic pain involves PB hyperactivity.
Collapse
Affiliation(s)
- Olivia Uddin
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States.,Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States
| | - Paige Studlack
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States.,Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States
| | - Titilola Akintola
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States
| | - Charles Raver
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States
| | - Alberto Castro
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States.,Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States
| | - Radi Masri
- Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, 650 W. Baltimore St, Baltimore, MD 21201, United States
| | - Asaf Keller
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States.,Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD 21201, United States
| |
Collapse
|
35
|
Peripheral 5-HT3 Receptors Are Involved in the Antinociceptive Effect of Bunodosine 391. Toxins (Basel) 2017; 10:toxins10010012. [PMID: 29280949 PMCID: PMC5793099 DOI: 10.3390/toxins10010012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/06/2017] [Accepted: 12/20/2017] [Indexed: 12/27/2022] Open
Abstract
Bunodosine 391 (BDS 391), a low molecular weight compound isolated from the sea anemone Bunodosoma cangicum, increases the nociceptive threshold and inhibits inflammatory hyperalgesia. Serotonin receptors are involved in those effects. In this study, we have expanded the characterization of the antinociceptive effect of BDS 391 demonstrating that, in rats: (a) the compound inhibits (1.2–12 ng/paw) overt pain, in the formalin test, and mechanical hyperalgesia (0.6–6.0 ng/paw) detected in a model of neuropathic pain; (b) intraplantar administration of ondansetron, a selective 5-HT3 receptor antagonist, blocks the effect of BDS 391, whereas ketanserin, a 5-HT2 receptor antagonist, partially reversed this effect, indicating the involvement of peripheral 5-HT2 and 5-HT3 receptors in BDS 391 antinociception; and (c) in binding assay studies, BDS 391 was not able to displace the selective 5-HT receptor antagonists, suggesting that this compound does not directly bind to these receptors. The effect of biguanide, a selective 5-HT3 receptor agonist, was also evaluated. The agonist inhibited the formalin’s nociceptive response, supporting an antinociceptive role for 5-HT3 receptors. Our study is the first one to show that a non-peptidic low molecular weight compound obtained from a sea anemone is able to induce antinociception and that activation of peripheral 5-HT3 receptors contributes to this effect.
Collapse
|
36
|
Akintola T, Raver C, Studlack P, Uddin O, Masri R, Keller A. The grimace scale reliably assesses chronic pain in a rodent model of trigeminal neuropathic pain. NEUROBIOLOGY OF PAIN 2017; 2:13-17. [PMID: 29450305 PMCID: PMC5808980 DOI: 10.1016/j.ynpai.2017.10.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Facial expressions were analyzed after constriction injury of infraorbital nerve. The grimace score reliably assesses ongoing pain in a this model. The grimace score can be used in both rats and mice with trigeminal neuropathic pain.
The limited success in translating basic science findings into effective pain management therapies reflects, in part, the difficulty in reliably assessing pain in experimental animals. This shortcoming is particularly acute in the field of chronic, ongoing pain. Quantitative analysis of facial expressions—the grimace score—was introduced as a promising tool, however, it is thought to reliably assess only pain of short or medium duration (minutes to hours). Here, we test the hypothesis that grimace scores are a reliable metric of ongoing neuropathic pain, by testing the prediction that chronic constriction injury of the infraorbital nerve (CCI-ION) will evoke significant increases in grimace scale scores. Mice and rats were subjected to CCI-ION, and tested for changes in mechanical hypersensitivity and in grimace scores, 10 or more days after surgery. Both rats and mice with CCI-ION had significantly higher grimace scores, and significantly lower thresholds for withdrawal from mechanical stimuli applied to the face, compared to sham-operated animals. Fentanyl reversed the changes in rat grimace scale scores, suggesting that these scores reflect pain perception. These findings validate the grimace scale as a reliable and sensitive metric for the assessment of ongoing pain in a rodent model of chronic, trigeminal neuropathic pain.
Collapse
Affiliation(s)
- Titilola Akintola
- Program in Toxicology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Charles Raver
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Paige Studlack
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Olivia Uddin
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Radi Masri
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Advanced Oral Sciences and Therapeutics, School of Dentistry, Baltimore, MD, USA
| | - Asaf Keller
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Sokolov AY, Sivachenko IB, Panteleev SS, Lyubashina OA. Blockade of 5-HT3 receptors with granisetron does not affect trigeminothalamic nociceptive transmission in rats: Implication for migraine. Clin Exp Pharmacol Physiol 2017; 45:34-41. [PMID: 28853174 DOI: 10.1111/1440-1681.12849] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/14/2017] [Accepted: 08/17/2017] [Indexed: 01/12/2023]
Abstract
One way to expand the existing range of anti-migraine drugs seems to be the search for pharmacological agents with anti-cephalalgic properties among medicines approved for clinical indications other than migraine. Numerous experimental and clinical data imply that selective serotonin 5-HT3 receptor antagonists can be considered as potential anti-migraine agents. Therefore, the objective of our work was to examine the impact of selective 5-HT3 receptor blockade with granisetron on migraine-related nociceptive transmission within the spinal trigeminal nucleus (STN) and the ventroposteromedial nucleus of the thalamus (VPM). Using an electrophysiological model of trigemino-durovascular nociception in anaesthetised male Wistar rats, we evaluated the effects of intravenous administration of granisetron on ongoing firing and dural electrical stimulation-evoked responses of the spinal trigeminal and thalamic cells. Granisetron did not substantially affect responses of the STN and VPM neurons to electrical stimulation of the dura mater as well as did not cause steady changes in ongoing firing of the spinal trigeminal cells. The results obtained argue against the use of 5-HT3 receptor antagonists for treating migraine. These data also lead to the conclusion that in the absence of sustained sensitisation of neurons along the trigemino-thalamo-cortical pathway the role of 5-HT3 receptor-dependent mechanisms in serotonergic modulation of trigeminovascular nociceptive transmission can hardly be considered crucial.
Collapse
Affiliation(s)
- Alexey Y Sokolov
- Department of Neuropharmacology, Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.,Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Ivan B Sivachenko
- Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Sergey S Panteleev
- Department of Neuropharmacology, Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.,Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Olga A Lyubashina
- Department of Neuropharmacology, Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, St. Petersburg, Russia.,Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
38
|
Castro A, Li Y, Raver C, Chandra R, Masri R, Lobo MK, Keller A. Neuropathic pain after chronic nerve constriction may not correlate with chloride dysregulation in mouse trigeminal nucleus caudalis neurons. Pain 2017; 158:1366-1372. [PMID: 28426550 PMCID: PMC5482239 DOI: 10.1097/j.pain.0000000000000926] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Changes in chloride reversal potential in rat spinal cord neurons have previously been associated with persistent pain in nerve injury and inflammation models. These changes correlate with a decrease in the expression of the potassium chloride transporter, KCC2, and with increases in neuronal excitability. Here, we test the hypothesis that similar changes occur in mice with neuropathic pain induced by chronic constriction injury of the trigeminal infraorbital nerve (CCI-ION). This model allows us to distinguish an acute pain phase (3-5 days after injury) from a persistent pain phase (12-14 days after CCI-ION). Chronic constriction injury of the trigeminal infraorbital nerve induced significant decreases in mechanical pain thresholds in both the acute and persistent phases. To estimate GABAA reversal potentials in neurons from trigeminal nucleus caudalis, we obtained perforated patch recordings in vitro. GABAA reversal potential decreased by 8% during the acute phase in unidentified neurons, but not in GABAergic interneurons. However, at 12 to 14 days after CCI-ION, GABAA reversal potential recovered to normal values. Quantitative real-time polymerase chain reaction analysis revealed no significant changes, at either 3 to 5 days or 12 to 14 days after CCI-ION, in either KCC2 or NKCC1. These findings suggest that CCI-ION in mice results in transient and modest changes in chloride reversal potentials, and that these changes may not persist during the late phase. This suggests that, in the mouse model of CCI-ION, chloride dysregulation may not have a prominent role in the central mechanisms leading to the maintenance of chronic pain.
Collapse
Affiliation(s)
- Alberto Castro
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine
- Program in Neuroscience, University of Maryland
| | - Ying Li
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine
- Program in Neuroscience, University of Maryland
| | - Charles Raver
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine
- Program in Neuroscience, University of Maryland
| | - Ramesh Chandra
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine
- Program in Neuroscience, University of Maryland
| | - Radi Masri
- Program in Neuroscience, University of Maryland
- Department of Endodontics, Prosthodontics and Operative Surgery, Baltimore College of Dentistry. Baltimore, MD 21201
| | - Mary Kay Lobo
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine
- Program in Neuroscience, University of Maryland
| | - Asaf Keller
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine
- Program in Neuroscience, University of Maryland
| |
Collapse
|
39
|
The developmental emergence of differential brainstem serotonergic control of the sensory spinal cord. Sci Rep 2017; 7:2215. [PMID: 28533557 PMCID: PMC5440407 DOI: 10.1038/s41598-017-02509-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/12/2017] [Indexed: 12/29/2022] Open
Abstract
Descending connections from brainstem nuclei are known to exert powerful control of spinal nociception and pain behaviours in adult mammals. Here we present evidence that descending serotonergic fibres not only inhibit nociceptive activity, but also facilitate non-noxious tactile activity in the healthy adult rat spinal dorsal horn via activation of spinal 5-HT3 receptors (5-HT3Rs). We further show that this differential serotonergic control in the adult emerges from a non-modality selective system in young rats. Serotonergic fibres exert background 5-HT3R mediated facilitation of both tactile and nociceptive spinal activity in the first three postnatal weeks. Thus, differential descending serotonergic control of spinal touch and pain processing emerges in late postnatal life to allow flexible and context-dependent brain control of somatosensation.
Collapse
|
40
|
Postnatal maturation of the spinal-bulbo-spinal loop: brainstem control of spinal nociception is independent of sensory input in neonatal rats. Pain 2016; 157:677-686. [PMID: 26574823 PMCID: PMC4751743 DOI: 10.1097/j.pain.0000000000000420] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The rostroventral medial medulla (RVM) is part of a rapidly acting spino-bulbo-spinal loop that is activated by ascending nociceptive inputs and drives descending feedback modulation of spinal nociception. In the adult rat, the RVM can facilitate or inhibit dorsal horn neuron inputs but in young animals descending facilitation dominates. It is not known whether this early life facilitation is part of a feedback loop. We hypothesized that the newborn RVM functions independently of sensory input, before the maturation of feedback control. We show here that noxious hind paw pinch evokes no fos activation in the RVM or the periaqueductal gray at postnatal day (P) 4 or P8, indicating a lack of nociceptive input at these ages. Significant fos activation was evident at P12, P21, and in adults. Furthermore, direct excitation of RVM neurons with microinjection of DL-homocysteic acid did not alter the net activity of dorsal horn neurons at P10, suggesting an absence of glutamatergic drive, whereas the same injections caused significant facilitation at P21. In contrast, silencing RVM neurons at P8 with microinjection of lidocaine inhibited dorsal horn neuron activity, indicating a tonic descending spinal facilitation from the RVM at this age. The results support the hypothesis that early life descending facilitation of spinal nociception is independent of sensory input. Since it is not altered by RVM glutamatergic receptor activation, it is likely generated by spontaneous brainstem activity. Only later in postnatal life can this descending activity be modulated by ascending nociceptive inputs in a functional spinal-bulbo-spinal loop.
Collapse
|
41
|
Mendonça MD, Caetano A, Viana-Baptista M. Association of depressive symptoms with allodynia in patients with migraine: A cross-sectional study. Cephalalgia 2016; 36:1077-1081. [DOI: 10.1177/0333102415620285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Aims Migraine and depression have a strong association. We aimed to determine whether this relationship was particularly evident in migraineurs with allodynia. Methods A cross-sectional study was carried out of 98 consecutive patients with episodic migraine presenting for their first evaluation in an outpatient clinic. The participants completed a demographic questionnaire, the Allodynia Symptom Checklist and the Hospital Anxiety and Depression Scale (HADS). Results Among the migraineurs, 75 (77%) reported allodynia. Allodynia was associated with higher median HADS-Anxiety (9 vs. 6, p = 0.038) and HADS-Depression (6 vs. 4, p = 0.014) scores. In a multiple regression model, the HADS-Depression scores were independently associated with allodynia (odds ratio 1.236, 95% confidence interval 1.046–1.461). An increased severity of allodynia correlated with higher depression scores ( r = 0.224; p = 0.027). Conclusion Anxious and depressive symptoms are more common in migraineurs with allodynia than in those without allodynia. Further studies are necessary to clarify the relationship between depressive symptoms and allodynia, as well as its therapeutic implications in migraine.
Collapse
Affiliation(s)
- Marcelo D Mendonça
- Neurology Department, Hospital Egas Moniz – Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- CEDOC, Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - André Caetano
- Neurology Department, Hospital Egas Moniz – Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Miguel Viana-Baptista
- Neurology Department, Hospital Egas Moniz – Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- CEDOC, Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
42
|
Mustafa G, Hou J, Tsuda S, Nelson R, Sinharoy A, Wilkie Z, Pandey R, Caudle RM, Neubert JK, Thompson FJ, Bose P. Trigeminal neuroplasticity underlies allodynia in a preclinical model of mild closed head traumatic brain injury (cTBI). Neuropharmacology 2016; 107:27-39. [PMID: 26972829 DOI: 10.1016/j.neuropharm.2016.03.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 01/10/2023]
Abstract
Post-traumatic headache (PTH) following TBI is a common and often persisting pain disability. PTH is often associated with a multimodal central pain sensitization on the skin surface described as allodynia. However, the particular neurobiology underlying cTBI-induced pain disorders are not known. These studies were performed to assess trigeminal sensory sensitization and to determine if sensitization measured behaviorally correlated with detectable changes in portions of the trigeminal sensory system (TSS), particularly trigeminal nucleus, thalamus, and sensory cortex. Thermal stimulation is particularly well suited to evaluate sensitization and was used in these studies. Recent advances in the use of reward/conflict paradigms permit use of operant measures of behavior, versus reflex-driven response behaviors, for thermal sensitization studies. Thus, to quantitate facial thermal sensitization (allodynia) in the setting of acute TBI, the current study utilized an operant orofacial pain reward/conflict testing paradigm to assess facial thermal sensitivity in uninjured control animals compared with those two weeks after cTBI in a rodent model. Significant reductions in facial contact/lick behaviors were observed in the TBI animals using either cool or warm challenge temperatures compared with behaviors in the normal animals. These facial thermal sensitizations correlated with detectable changes in multiple levels of the TSS. The immunohistochemical (IHC) studies revealed significant alterations in the expression of the serotonin (5-HT), neurokinin 1 receptor (NK1R), norepinephrine (NE), and gamma-aminobutyric acid (GABA) in the caudal trigeminal nucleus, thalamic VPL/VPM nucleus, and sensory cortex of the orofacial pain pathways. There was a strong correlation between increased expression of certain IHC markers and increased behavioral markers for facial sensitization. The authors conclude that TBI-induced changes observed in the TSS are consistent with the expression of generalized facial allodynia following cTBI. To our knowledge, this is the first report of orofacial sensitization correlated with changes in selected neuromodulators/neurotransmitters in the TSS following experimental mild TBI.
Collapse
Affiliation(s)
- Golam Mustafa
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA; Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0144, USA
| | - Jiamei Hou
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA; Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0144, USA
| | - Shigeharu Tsuda
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0144, USA
| | - Rachel Nelson
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA
| | - Ankita Sinharoy
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA
| | - Zachary Wilkie
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA
| | - Rahul Pandey
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA
| | - Robert M Caudle
- Department of Oral and Maxillofacial Surgery, College of Dentistry, University of Florida, Gainesville, FL 32610-0244, USA
| | - John K Neubert
- Department of Orthodontics, College of Dentistry, University of Florida, Gainesville, FL 32610-0244, USA
| | - Floyd J Thompson
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA; Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0144, USA; Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610-0244, USA
| | - Prodip Bose
- Brain Rehabilitation Research Center of Excellence, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, Gainesville, FL 32608-1197, USA; Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0144, USA; Department of Neurology, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA.
| |
Collapse
|
43
|
Dieb W, Ouachikh O, Alves S, Boucher Y, Durif F, Hafidi A. Nigrostriatal dopaminergic depletion increases static orofacial allodynia. J Headache Pain 2016; 17:11. [PMID: 26885825 PMCID: PMC4757596 DOI: 10.1186/s10194-016-0607-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/12/2016] [Indexed: 01/05/2023] Open
Abstract
Background This study investigated mesencephalic dopamine depletion effects on static mechanical allodynia (SMA) elicited by chronic constriction of the infraorbitary nerve (CCI-IoN). Methods Dopamine depletion (6-OHDA administration into the medial forebrain bundle) effects on CCI-IoN-induced SMA were explored using behavioral (nocifensive behavior score upon non-noxious stimuli using von Frey filament), pharmacological (bromocriptine injections) and immunohistochemical (PKCγ and pERK1/2) techniques. Results The central dopamine depletion increased significantly the SMA score. Intraperitoneal and intracisternal injections of bromocriptine alleviated the allodynic behavior observed in both CCI-IoN and CCI-IoN + 6-OHDA animal groups. At the cellular level, dopamine depletion induced a significant increase in PKCγ expression in the medullary dorsal horn (MDH) in rat with CCI-IoN + 6-OHDA when compared to sham animals (CCI-IoN only). Similarly, after static non-noxious stimuli, the expression of pain marker proteins pERK1/2 within the MDH revealed significantly a higher number of positive cells in CCI-IoN + 6-OHDA rats when compared to the CCI-IoN group. Conclusion This study demonstrates that nigrostriatal dopamine depletion exacerbates the neuropathic pain resulting from CCI-IoN. This effect is probably due to an action through descending pain inhibitory systems which increased pain sensitization at the MDH level. It demonstrates also an analgesic effect elicited by D2R activation at the segmental level. Electronic supplementary material The online version of this article (doi:10.1186/s10194-016-0607-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wisam Dieb
- UFR Odontologie, Université Paris Diderot, Paris, France. .,Centre de Psychiatrie et Neurosciences, INSERM U894, Paris, France. .,Clermont Université, Université d'Auvergne, EA7280, Clermont-Ferrand, France.
| | - Omar Ouachikh
- Clermont Université, Université d'Auvergne, EA7280, Clermont-Ferrand, France.
| | - Sofia Alves
- Clermont Université, Université d'Auvergne, EA7280, Clermont-Ferrand, France.
| | - Yves Boucher
- UFR Odontologie, Université Paris Diderot, Paris, France. .,Centre de Psychiatrie et Neurosciences, INSERM U894, Paris, France.
| | - Franck Durif
- Clermont Université, Université d'Auvergne, EA7280, Clermont-Ferrand, France. .,CHU Clermont-Ferrand, Service de Neurologie, 63000, Clermont-Ferrand, France.
| | - Aziz Hafidi
- Clermont Université, Université d'Auvergne, EA7280, Clermont-Ferrand, France.
| |
Collapse
|
44
|
Emerging Role of Spinal Cord TRPV1 in Pain Exacerbation. Neural Plast 2016; 2016:5954890. [PMID: 26885404 PMCID: PMC4738952 DOI: 10.1155/2016/5954890] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/20/2015] [Accepted: 08/12/2015] [Indexed: 12/25/2022] Open
Abstract
TRPV1 is well known as a sensor ion channel that transduces a potentially harmful environment into electrical depolarization of the peripheral terminal of the nociceptive primary afferents. Although TRPV1 is also expressed in central regions of the nervous system, its roles in the area remain unclear. A series of recent reports on the spinal cord synapses have provided evidence that TRPV1 plays an important role in synaptic transmission in the pain pathway. Particularly, in pathologic pain states, TRPV1 in the central terminal of sensory neurons and interneurons is suggested to commonly contribute to pain exacerbation. These observations may lead to insights regarding novel synaptic mechanisms revealing veiled roles of spinal cord TRPV1 and may offer another opportunity to modulate pathological pain by controlling TRPV1. In this review, we introduce historical perspectives of this view and details of the recent promising results. We also focus on extended issues and unsolved problems to fully understand the role of TRPV1 in pathological pain. Together with recent findings, further efforts for fine analysis of TRPV1's plastic roles in pain synapses at different levels in the central nervous system will promote a better understanding of pathologic pain mechanisms and assist in developing novel analgesic strategies.
Collapse
|
45
|
Wang J, Li ZH, Feng B, Zhang T, Zhang H, Li H, Chen T, Cui J, Zang WD, Li YQ. Corticotrigeminal Projections from the Insular Cortex to the Trigeminal Caudal Subnucleus Regulate Orofacial Pain after Nerve Injury via Extracellular Signal-Regulated Kinase Activation in Insular Cortex Neurons. Front Cell Neurosci 2015; 9:493. [PMID: 26733817 PMCID: PMC4689789 DOI: 10.3389/fncel.2015.00493] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 12/08/2015] [Indexed: 11/16/2022] Open
Abstract
Cortical neuroplasticity alterations are implicated in the pathophysiology of chronic orofacial pain. However, the relationship between critical cortex excitability and orofacial pain maintenance has not been fully elucidated. We recently demonstrated a top-down corticospinal descending pain modulation pathway from the anterior cingulate cortex (ACC) to the spinal dorsal horn that could directly regulate nociceptive transmission. Thus, we aimed to investigate possible corticotrigeminal connections that directly influence orofacial nociception in rats. Infraorbital nerve chronic constriction injury (IoN-CCI) induced significant orofacial nociceptive behaviors as well as pain-related negative emotions such as anxiety/depression in rats. By combining retrograde and anterograde tract tracing, we found powerful evidence that the trigeminal caudal subnucleus (Vc), especially the superficial laminae (I/II), received direct descending projections from granular and dysgranular parts of the insular cortex (IC). Extracellular signal-regulated kinase (ERK), an important signaling molecule involved in neuroplasticity, was significantly activated in the IC following IoN-CCI. Moreover, in IC slices from IoN-CCI rats, U0126, an inhibitor of ERK activation, decreased both the amplitude and the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) and reduced the paired-pulse ratio (PPR) of Vc-projecting neurons. Additionally, U0126 also reduced the number of action potentials in the Vc-projecting neurons. Finally, intra-IC infusion of U0126 obviously decreased Fos expression in the Vc, accompanied by the alleviation of both nociceptive behavior and negative emotions. Thus, the corticotrigeminal descending pathway from the IC to the Vc could directly regulate orofacial pain, and ERK deactivation in the IC could effectively alleviate neuropathic pain as well as pain-related negative emotions in IoN-CCI rats, probably through this top–down pathway. These findings may help researchers and clinicians to better understand the underlying modulation mechanisms of orofacial neuropathic pain and indicate a novel mechanism of ERK inhibitor-induced analgesia.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Zhi-Hua Li
- Basic Medical College, Zhengzhou University Zhengzhou, China
| | - Ban Feng
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Ting Zhang
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Han Zhang
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Hui Li
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Tao Chen
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Jing Cui
- Basic Medical College, Zhengzhou University Zhengzhou, China
| | - Wei-Dong Zang
- Basic Medical College, Zhengzhou University Zhengzhou, China
| | - Yun-Qing Li
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical UniversityXi'an, China; Collaborative Innovation Center for Brain Science, Fudan UniversityShanghai, China
| |
Collapse
|
46
|
Galor A, Covington D, Levitt AE, McManus KT, Seiden B, Felix ER, Kalangara J, Feuer W, Patin DJ, Martin ER, Sarantopoulos KD, Levitt RC. Neuropathic Ocular Pain due to Dry Eye is Associated with Multiple Comorbid Chronic Pain Syndromes. THE JOURNAL OF PAIN 2015; 17:310-8. [PMID: 26606863 DOI: 10.1016/j.jpain.2015.10.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/19/2015] [Accepted: 10/29/2015] [Indexed: 12/30/2022]
Abstract
UNLABELLED Recent data show that dry eye (DE) susceptibility and other chronic pain syndromes (CPS) such as chronic widespread pain, irritable bowel syndrome, and pelvic pain, might share common heritable factors. Previously, we showed that DE patients described more severe symptoms and tended to report features of neuropathic ocular pain (NOP). We hypothesized that patients with a greater number of CPS would have a different DE phenotype compared with those with fewer CPS. We recruited a cohort of 154 DE patients from the Miami Veterans Affairs Hospital and defined high and low CPS groups using cluster analysis. In addition to worse nonocular pain complaints and higher post-traumatic stress disorder and depression scores (P < .01), we found that the high CPS group reported more severe neuropathic type DE symptoms compared with the low CPS group, including worse ocular pain assessed via 3 different pain scales (P < .05), with similar objective corneal DE signs. To our knowledge, this was the first study to show that DE patients who manifest a greater number of comorbid CPS reported more severe DE symptoms and features of NOP. These findings provided further evidence that NOP might represent a central pain disorder, and that shared mechanistic factors might underlie vulnerability to some forms of DE and other comorbid CPS. PERSPECTIVE DE patients reported more frequent CPS (high CPS group) and reported worse DE symptoms and ocular and nonocular pain scores. The high CPS group reported symptoms of NOP that share causal genetic factors with comorbid CPS. These results imply that an NOP evaluation and treatment should be considered for DE patients.
Collapse
Affiliation(s)
- Anat Galor
- Miami Veterans Administration Medical Center, Miami, Florida; Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | - Derek Covington
- Miami Veterans Administration Medical Center, Miami, Florida; Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami Miller School of Medicine, Miami, Florida
| | | | - Katherine T McManus
- Miami Veterans Administration Medical Center, Miami, Florida; Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | - Benjamin Seiden
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | - Elizabeth R Felix
- Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami Miller School of Medicine, Miami, Florida; Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida
| | - Jerry Kalangara
- Miami Veterans Administration Medical Center, Miami, Florida; Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami Miller School of Medicine, Miami, Florida
| | - William Feuer
- Bascom Palmer Eye Institute, University of Miami, Miami, Florida
| | - Dennis J Patin
- Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami Miller School of Medicine, Miami, Florida
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida; John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Konstantinos D Sarantopoulos
- Miami Veterans Administration Medical Center, Miami, Florida; Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami Miller School of Medicine, Miami, Florida
| | - Roy C Levitt
- Miami Veterans Administration Medical Center, Miami, Florida; Department of Anesthesiology, Perioperative Medicine and Pain Management, University of Miami Miller School of Medicine, Miami, Florida; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida; John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
47
|
Activity-triggered tetrapartite neuron-glial interactions following peripheral injury. Curr Opin Pharmacol 2015; 26:16-25. [PMID: 26431645 DOI: 10.1016/j.coph.2015.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/14/2015] [Accepted: 09/16/2015] [Indexed: 12/11/2022]
Abstract
Recent studies continue to support the proposition that non-neuronal components of the nervous system, mainly glial cells and associated chemical mediators, contribute to the development of neuronal hyperexcitability that underlies persistent pain conditions. In the event of peripheral injury, enhanced or abnormal nerve input is likely the most efficient way to activate simultaneously central neurons and glia. Injury induces phenotypic changes in glia and triggers signaling cascades that engage reciprocal interactions between presynaptic terminals, postsynaptic neurons, microglia and astrocytes. While some responses to peripheral injury may help the nervous system to adapt positively to counter the disastrous effect of injury, the net effect often leads to long-lasting sensitization of pain transmission pathways and chronic pain.
Collapse
|
48
|
Smith JB, Watson GDR, Alloway KD, Schwarz C, Chakrabarti S. Corticofugal projection patterns of whisker sensorimotor cortex to the sensory trigeminal nuclei. Front Neural Circuits 2015; 9:53. [PMID: 26483640 PMCID: PMC4588702 DOI: 10.3389/fncir.2015.00053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/14/2015] [Indexed: 11/29/2022] Open
Abstract
The primary (S1) and secondary (S2) somatosensory cortices project to several trigeminal sensory nuclei. One putative function of these corticofugal projections is the gating of sensory transmission through the trigeminal principal nucleus (Pr5), and some have proposed that S1 and S2 project differentially to the spinal trigeminal subnuclei, which have inhibitory circuits that could inhibit or disinhibit the output projections of Pr5. Very little, however, is known about the origin of sensorimotor corticofugal projections and their patterns of termination in the various trigeminal nuclei. We addressed this issue by injecting anterograde tracers in S1, S2 and primary motor (M1) cortices, and quantitatively characterizing the distribution of labeled terminals within the entire rostro-caudal chain of trigeminal sub-nuclei. We confirmed our anterograde tracing results by injecting retrograde tracers at various rostro-caudal levels within the trigeminal sensory nuclei to determine the position of retrogradely labeled cortical cells with respect to S1 barrel cortex. Our results demonstrate that S1 and S2 projections terminate in largely overlapping regions but show some significant differences. Whereas S1 projection terminals tend to cluster within the principal trigeminal (Pr5), caudal spinal trigeminal interpolaris (Sp5ic), and the dorsal spinal trigeminal caudalis (Sp5c), S2 projection terminals are distributed in a continuum across all trigeminal nuclei. Contrary to the view that sensory gating could be mediated by differential activation of inhibitory interconnections between the spinal trigeminal subnuclei, we observed that projections from S1 and S2 are largely overlapping in these subnuclei despite the differences noted earlier.
Collapse
Affiliation(s)
- Jared B Smith
- Department of Engineering Science and Mechanics, Pennsylvania State University University Park, PA, USA ; Center for Neural Engineering, Huck Institute of Life Sciences, Pennsylvania State University University Park, PA, USA
| | - Glenn D R Watson
- Center for Neural Engineering, Huck Institute of Life Sciences, Pennsylvania State University University Park, PA, USA ; Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine Hershey, PA, USA
| | - Kevin D Alloway
- Center for Neural Engineering, Huck Institute of Life Sciences, Pennsylvania State University University Park, PA, USA ; Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine Hershey, PA, USA
| | - Cornelius Schwarz
- Department of Cognitive Neurology, Hertie Institute for Clinical Brain Research, Eberhard Karls University of Tübingen Tübingen, Germany ; Systems Neurophysiology, Werner Reichardt Center for Integrative Neurosciences, Eberhard Karls University of Tübingen Tübingen, Germany
| | - Shubhodeep Chakrabarti
- Department of Cognitive Neurology, Hertie Institute for Clinical Brain Research, Eberhard Karls University of Tübingen Tübingen, Germany ; Systems Neurophysiology, Werner Reichardt Center for Integrative Neurosciences, Eberhard Karls University of Tübingen Tübingen, Germany
| |
Collapse
|
49
|
Okamoto K, Katagiri A, Rahman M, Thompson R, Bereiter DA. Inhibition of temporomandibular joint input to medullary dorsal horn neurons by 5HT3 receptor antagonist in female rats. Neuroscience 2015; 299:35-44. [PMID: 25913635 DOI: 10.1016/j.neuroscience.2015.04.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/17/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022]
Abstract
Repeated forced swim (FS) conditioning enhances nociceptive responses to temporomandibular joint (TMJ) stimulation in female rats. The basis for FS-induced TMJ hyperalgesia remains unclear. To test the hypothesis that serotonin 3 receptor (5HT3R) mechanisms contribute to enhanced TMJ nociception after FS, ovariectomized female rats were treated with estradiol and subjected to FS for three days. On day 4, rats were anesthetized with isoflurane and TMJ-responsive neurons were recorded from superficial and deep laminae at the trigeminal subnucleus caudalis/upper cervical (Vc/C1-2) region and electromyographic (EMG) activity was recorded from the masseter muscle. Only Vc/C1-2 neurons activated by intra-TMJ injections of ATP were included for further analysis. Although neurons in both superficial and deep laminae were activated by ATP, only neurons in deep laminae displayed enhanced responses after FS. Local application of the 5HT3R antagonist, ondansetron (OND), at the Vc/C1-2 region reduced the ATP-evoked responses of neurons in superficial and deep laminae and reduced the EMG response in both sham and FS rats. OND also decreased the spontaneous firing rate of neurons in deep laminae and reduced the high-threshold convergent cutaneous receptive field area of neurons in superficial and deep laminae in both sham and FS rats. These results revealed that central application of a 5HT3R antagonist, had widespread effects on the properties of TMJ-responsive neurons at the Vc/C1-2 region and on jaw muscle reflexes under sham and FS conditions. It is concluded that 5HT3R does not play a unique role in mediating stress-induced hyperalgesia related to TMJ nociception.
Collapse
Affiliation(s)
- K Okamoto
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, United States.
| | - A Katagiri
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, United States
| | - M Rahman
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, United States
| | - R Thompson
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, United States
| | - D A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, United States
| |
Collapse
|
50
|
Neuroplasticity underlying the comorbidity of pain and depression. Neural Plast 2015; 2015:504691. [PMID: 25810926 PMCID: PMC4355564 DOI: 10.1155/2015/504691] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/10/2015] [Indexed: 02/07/2023] Open
Abstract
Acute pain induces depressed mood, and chronic pain is known to cause depression. Depression, meanwhile, can also adversely affect pain behaviors ranging from symptomology to treatment response. Pain and depression independently induce long-term plasticity in the central nervous system (CNS). Comorbid conditions, however, have distinct patterns of neural activation. We performed a review of the changes in neural circuitry and molecular signaling pathways that may underlie this complex relationship between pain and depression. We also discussed some of the current and future therapies that are based on this understanding of the CNS plasticity that occurs with pain and depression.
Collapse
|