1
|
Katagiri A, Kishimoto S, Okamoto Y, Yamada M, Niwa H, Bereiter DA, Kato T. Effect of chronic intermittent hypoxia on ocular and intraoral mechanical allodynia mediated via the calcitonin gene-related peptide in a rat. Sleep 2024; 47:zsad332. [PMID: 38166171 PMCID: PMC10925949 DOI: 10.1093/sleep/zsad332] [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: 08/09/2023] [Revised: 12/15/2023] [Indexed: 01/04/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea, a significant hypoxic condition, may exacerbate several orofacial pain conditions. The study aims to define the involvement of calcitonin gene-related peptide (CGRP) in peripheral and central sensitization and in evoking orofacial mechanical allodynia under chronic intermittent hypoxia (CIH). METHODS Male rats were exposed to CIH. Orofacial mechanical allodynia was assessed using the eyeblink test and the two-bottle preference drinking test. The CGRP-immunoreactive neurons in the trigeminal ganglion (TG), CGRP-positive primary afferents projecting to laminae I-II of the trigeminal spinal subnucleus caudalis (Vc), and neural responses in the second-order neurons of the Vc were determined by immunohistochemistry. CGRP receptor antagonist was administrated in the TG. RESULTS CIH-induced ocular and intraoral mechanical allodynia. CGRP-immunoreactive neurons and activated satellite glial cells (SGCs) were significantly increased in the TG and the number of cFos-immunoreactive cells in laminae I-II of the Vc were significantly higher in CIH rats compared to normoxic rats. Local administration of the CGRP receptor antagonist in the TG of CIH rats attenuated orofacial mechanical allodynia; the number of CGRP-immunoreactive neurons and activated SGCs in the TG, and the density of CGRP-positive primary afferent terminals and the number of cFos-immunoreactive cells in laminae I-II of the Vc were significantly lower compared to vehicle-administrated CIH rats. CONCLUSIONS An increase in CGRP in the TG induced by CIH, as well as orofacial mechanical allodynia and central sensitization of second-order neurons in the Vc, supported the notion that CGRP plays a critical role in CIH-induced orofacial mechanical allodynia.
Collapse
Affiliation(s)
- Ayano Katagiri
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Saki Kishimoto
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yoshie Okamoto
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masaharu Yamada
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hitoshi Niwa
- Department of Dental Anesthesiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - David A Bereiter
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, MN, USA
| | - Takafumi Kato
- Department of Oral Physiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
2
|
Pathophysiology of Post-Traumatic Trigeminal Neuropathic Pain. Biomolecules 2022; 12:biom12121753. [PMID: 36551181 PMCID: PMC9775491 DOI: 10.3390/biom12121753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/29/2022] Open
Abstract
Trigeminal nerve injury is one of the causes of chronic orofacial pain. Patients suffering from this condition have a significantly reduced quality of life. The currently available management modalities are associated with limited success. This article reviews some of the common causes and clinical features associated with post-traumatic trigeminal neuropathic pain (PTNP). A cascade of events in the peripheral and central nervous system function is involved in the pathophysiology of pain following nerve injuries. Central and peripheral processes occur in tandem and may often be co-dependent. Due to the complexity of central mechanisms, only peripheral events contributing to the pathophysiology have been reviewed in this article. Future investigations will hopefully help gain insight into trigeminal-specific events in the pathophysiology of the development and maintenance of neuropathic pain secondary to nerve injury and enable the development of new therapeutic modalities.
Collapse
|
3
|
Reedich EJ, Genry LT, Singer MA, Cavarsan CF, Mena Avila E, Boudreau DM, Brennan MC, Garrett AM, Dowaliby L, Detloff MR, Quinlan KA. Enhanced nociceptive behavior and expansion of associated primary afferents in a rabbit model of cerebral palsy. J Neurosci Res 2022; 100:1951-1966. [PMID: 35839339 PMCID: PMC9388620 DOI: 10.1002/jnr.25108] [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: 01/03/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 11/07/2022]
Abstract
Spastic cerebral palsy (CP) is a movement disorder marked by hypertonia and hyperreflexia; the most prevalent comorbidity is pain. Since spinal nociceptive afferents contribute to both the sensation of painful stimuli as well as reflex circuits involved in movement, we investigated the relationship between prenatal hypoxia-ischemia (HI) injury which can cause CP, and possible changes in spinal nociceptive circuitry. To do this, we examined nociceptive afferents and mechanical and thermal sensitivity of New Zealand White rabbit kits after prenatal HI or a sham surgical procedure. As described previously, a range of motor deficits similar to spastic CP was observed in kits born naturally after HI (40 min at ~70%-80% gestation). We found that HI caused an expansion of peptidergic afferents (marked by expression of calcitonin gene-related peptide) in both the superficial and deep dorsal horn at postnatal day (P)5. Non-peptidergic nociceptive afferent arborization (labeled by isolectin B4) was unaltered in HI kits, but overlap of the two populations (peptidergic and non-peptidergic nociceptors) was increased by HI. Density of glial fibrillary acidic protein was unchanged within spinal cord white matter regions important in nociceptive transmission at P5. We found that mechanical and thermal nociception was enhanced in HI kits even in the absence of motor deficits. These findings suggest that prenatal HI injury impacts spinal sensory pathways in addition to the more well-established disruptions to descending motor circuits. In conclusion, changes to spinal nociceptive circuitry could disrupt spinal reflexes and contribute to pain experienced by individuals with CP.
Collapse
Affiliation(s)
- Emily J Reedich
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Landon T Genry
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, Rhode Island, USA
| | - Meredith A Singer
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA
| | - Clarissa Fantin Cavarsan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Elvia Mena Avila
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Daphne M Boudreau
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Michael C Brennan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Alyssa M Garrett
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Rhode Island Institutional Development Award (IDeA) Network for Biomedical Research Excellence (INBRE) Summer Undergraduate Research Fellowship (SURF) Program, University of Rhode Island, Kingston, Rhode Island, USA
| | - Lisa Dowaliby
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - Megan R Detloff
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA
| | - Katharina A Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| |
Collapse
|
4
|
Ishida K, Tanaka S, Shen D, Matsui S, Fuseya S, Shindo T, Kawamata M. Calcitonin gene-related peptide is not involved in neuropathic pain induced by partial sciatic nerve ligation in mice. Neurosci Lett 2022; 778:136615. [DOI: 10.1016/j.neulet.2022.136615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022]
|
5
|
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
|
6
|
Meewis J, Renton T, Jacobs R, Politis C, Van der Cruyssen F. Post-traumatic trigeminal neuropathy: correlation between objective and subjective assessments and a prediction model for neurosensory recovery. J Headache Pain 2021; 22:44. [PMID: 34030632 PMCID: PMC8146662 DOI: 10.1186/s10194-021-01261-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Post-traumatic trigeminal neuropathy (PTN) can have a substantial effect on patient well-being. However, the relation between the neuropathic symptoms and their effect on psychosocial functioning remains a matter of debate. The purpose of this study was to evaluate the association between objective and subjective assessments of neurosensory function in PTN and predict neurosensory outcome using baseline measurements. METHODS This prospective observational cohort study included patients diagnosed with PTN at the Department of Oral and Maxillofacial Surgery, University Hospital Leuven, Belgium, between April 2018 and May 2020. Standardized objective and subjective neurosensory examinations were recorded simultaneously on multiple occasions during the follow-up period. Correlation analyses and principal component analysis were conducted, and a prediction model of neurosensory recovery was developed. RESULTS Quality of life correlated significantly (P < 0.05) with percentage of affected dermatome (ρ = - 0.35), the presence of brush stroke allodynia (ρ = - 0.24), gain-of-function sensory phenotype (ρ = - 0.41), Medical Research Council Scale (ρ = 0.36), and Sunderland classification (ρ = - 0.21). Quality of life was not significantly correlated (P > 0.05) with directional discrimination, stimulus localization, two-point discrimination, or sensory loss-of-function. The prediction model showed a negative predictive value for neurosensory recovery after 6 months of 87%. CONCLUSIONS We found a strong correlation of subjective well-being with the presence of brush stroke allodynia, thermal and/or mechanical hyperesthesia, and the size of the neuropathic area. These results suggest that positive symptoms dominate the effect on affect. In patients reporting poor subjective well-being in the absence of positive symptoms or a large neuropathic area, additional attention towards psychosocial triggers might enhance treatment outcome. The prediction model could contribute to establishing realistic expectations about the likelihood of neurosensory recovery but remains to be validated in future studies.
Collapse
Affiliation(s)
- Jeroen Meewis
- Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Kapucijnenvoer 33, 3000, Leuven, Belgium.
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, University Leuven, Leuven, Belgium.
| | - Tara Renton
- Department of Oral Surgery, King's College London Dental Institute, London, UK
| | - Reinhilde Jacobs
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, University Leuven, Leuven, Belgium
- Department of Dental Medicine, Karolinska institutet, Stockholm, Sweden
| | - Constantinus Politis
- Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Kapucijnenvoer 33, 3000, Leuven, Belgium
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, University Leuven, Leuven, Belgium
| | - Fréderic Van der Cruyssen
- Department of Oral and Maxillofacial Surgery, University Hospitals Leuven, Kapucijnenvoer 33, 3000, Leuven, Belgium
- OMFS-IMPATH Research Group, Department of Imaging and Pathology, Faculty of Medicine, University Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Kang SA, Govindarajan R. Anti-calcitonin gene-related peptide monoclonal antibodies for neuropathic pain in patients with migraine headache. Muscle Nerve 2021; 63:563-567. [PMID: 33347632 DOI: 10.1002/mus.27153] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/16/2020] [Accepted: 12/19/2020] [Indexed: 01/03/2023]
Abstract
INTRODUCTION There is increasing evidence that calcitonin gene-related peptide (CGRP) plays a role in the development of neuropathic pain, a common feature of peripheral neuropathy. Although clinical studies have shown that anti-CGRP monoclonal antibodies are highly efficacious for migraine headache prophylaxis, their effects on nonheadache chronic pain conditions, including neuropathic pain, in humans are unknown. Therefore, the aim of this study was to assess the effectiveness of anti-CGRP monoclonal antibodies for neuropathic pain in patients with coexisting chronic migraine. METHODS A retrospective chart review was conducted of 14 patients with chronic migraine and peripheral neuropathy. All patients were treated with anti-CGRP monoclonal antibodies. We collected data on patient-reported scores on the Neuropathy Pain Scale (NPS) and the frequency of migraine headache days (MHDs) per month. Data were collected 3 and 0 months before and 3, 6, 9, and 12 months after treatment with anti-CGRP medications. RESULTS With treatment of anti-CGRP monoclonal antibodies, patients reported a 41.7% decrease in NPS scores from 89.3 at baseline to 52.1 at 12 months posttreatment (P < .05). In addition, there was a 33.3% decrease in MHDs per month from 19.8 at baseline to 13.2 at 12 months posttreatment (P < .05). DISCUSSION Administration of anti-CGRP medications significantly improved neuropathic pain in patients who also had chronic migraine. To confirm these promising outcomes, it would be worthwhile to conduct a blinded, randomized study with a larger population of patients.
Collapse
Affiliation(s)
- Seung Ah Kang
- Department of Neurology, University of Missouri School of Medicine, Columbia, Missouri
| | - Raghav Govindarajan
- Department of Neurology, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
8
|
Bernal L, Cisneros E, Roza C. Activation of the regeneration-associated gene STAT3 and functional changes in intact nociceptors after peripheral nerve damage in mice. Eur J Pain 2021; 25:886-901. [PMID: 33345380 DOI: 10.1002/ejp.1718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND In the context of neuropathic pain, the contribution of regeneration to the development of positive symptoms is not completely understood. Several efforts have been done to described changes in axotomized neurons, however, there is scarce data on changes occurring in intact neurons, despite experimental evidence of functional changes. To address this issue, we analysed by immunohistochemistry the presence of phosphorylated signal transducer and activator of transcription 3 (pSTAT3), an accepted marker of regeneration, within DRGs where axotomized neurons were retrogradely labelled following peripheral nerve injury. Likewise, we have characterized abnormal electrophysiological properties in intact fibres after partial nerve injury. METHODS/RESULTS We showed that induction of pSTAT3 in sensory neurons was similar after partial or total transection of the sciatic nerve and to the same extent within axotomized and non-axotomized neurons. We also examined pSTAT3 presence on non-peptidergic and peptidergic nociceptors. Whereas the percentage of neurons marked by IB4 decrease after injury, the proportion of CGRP neurons did not change, but its expression switched from small- to large-diameter neurons. Besides, the percentage of CGRP+ neurons expressing pSTAT3 increased significantly 2.5-folds after axotomy, preferentially in neurons with large diameters. Electrophysiological recordings showed that after nerve damage, most of the neurons with ectopic spontaneous activity (39/46) were non-axotomized C-fibres with functional receptive fields in the skin far beyond the site of damage. CONCLUSIONS Neuronal regeneration after nerve injury, likely triggered from the site of injury, may explain the abnormal functional properties gained by intact neurons, reinforcing their role in neuropathic pain. SIGNIFICANCE Positive symptoms in patients with peripheral neuropathies correlate to abnormal functioning of different subpopulations of primary afferents. Peripheral nerve damage triggers regenerating programs in the cell bodies of axotomized but also in non-axotomized nociceptors which is in turn, develop abnormal spontaneous and evoked discharges. Therefore, intact nociceptors have a significant role in the development of neuropathic pain due to their hyperexcitable peripheral terminals. Therapeutical targets should focus on inhibiting peripheral hyperexcitability in an attempt to limit peripheral and central sensitization.
Collapse
Affiliation(s)
- Laura Bernal
- Department of System's Biology, Medical School, University of Alcala, Alcalá de Henares, Spain
| | - Elsa Cisneros
- Department of System's Biology, Medical School, University of Alcala, Alcalá de Henares, Spain.,Health Sciences School, Centro Universitario Internacional de Madrid (CUNIMAD), Madrid, Spain.,Health Sciences School, Universidad Internacional de La Rioja (UNIR), Logroño, Spain
| | - Carolina Roza
- Department of System's Biology, Medical School, University of Alcala, Alcalá de Henares, Spain
| |
Collapse
|
9
|
Tran EL, Crawford LK. Revisiting PNS Plasticity: How Uninjured Sensory Afferents Promote Neuropathic Pain. Front Cell Neurosci 2020; 14:612982. [PMID: 33362476 PMCID: PMC7759741 DOI: 10.3389/fncel.2020.612982] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/12/2020] [Indexed: 11/13/2022] Open
Abstract
Despite the widespread study of how injured nerves contribute to chronic pain, there are still major gaps in our understanding of pain mechanisms. This is particularly true of pain resulting from nerve injury, or neuropathic pain, wherein tactile or thermal stimuli cause painful responses that are particularly difficult to treat with existing therapies. Curiously, this stimulus-driven pain relies upon intact, uninjured sensory neurons that transmit the signals that are ultimately sensed as painful. Studies that interrogate uninjured neurons in search of cell-specific mechanisms have shown that nerve injury alters intact, uninjured neurons resulting in an activity that drives stimulus-evoked pain. This review of neuropathic pain mechanisms summarizes cell-type-specific pathology of uninjured sensory neurons and the sensory ganglia that house their cell bodies. Uninjured neurons have demonstrated a wide range of molecular and neurophysiologic changes, many of which are distinct from those detected in injured neurons. These intriguing findings include expression of pain-associated molecules, neurophysiological changes that underlie increased excitability, and evidence that intercellular signaling within sensory ganglia alters uninjured neurons. In addition to well-supported findings, this review also discusses potential mechanisms that remain poorly understood in the context of nerve injury. This review highlights key questions that will advance our understanding of the plasticity of sensory neuron subpopulations and clarify the role of uninjured neurons in developing anti-pain therapies.
Collapse
Affiliation(s)
- Emily L Tran
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| | - LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, United States
| |
Collapse
|
10
|
Smith PA. K + Channels in Primary Afferents and Their Role in Nerve Injury-Induced Pain. Front Cell Neurosci 2020; 14:566418. [PMID: 33093824 PMCID: PMC7528628 DOI: 10.3389/fncel.2020.566418] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Sensory abnormalities generated by nerve injury, peripheral neuropathy or disease are often expressed as neuropathic pain. This type of pain is frequently resistant to therapeutic intervention and may be intractable. Numerous studies have revealed the importance of enduring increases in primary afferent excitability and persistent spontaneous activity in the onset and maintenance of peripherally induced neuropathic pain. Some of this activity results from modulation, increased activity and /or expression of voltage-gated Na+ channels and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. K+ channels expressed in dorsal root ganglia (DRG) include delayed rectifiers (Kv1.1, 1.2), A-channels (Kv1.4, 3.3, 3.4, 4.1, 4.2, and 4.3), KCNQ or M-channels (Kv7.2, 7.3, 7.4, and 7.5), ATP-sensitive channels (KIR6.2), Ca2+-activated K+ channels (KCa1.1, 2.1, 2.2, 2.3, and 3.1), Na+-activated K+ channels (KCa4.1 and 4.2) and two pore domain leak channels (K2p; TWIK related channels). Function of all K+ channel types is reduced via a multiplicity of processes leading to altered expression and/or post-translational modification. This also increases excitability of DRG cell bodies and nociceptive free nerve endings, alters axonal conduction and increases neurotransmitter release from primary afferent terminals in the spinal dorsal horn. Correlation of these cellular changes with behavioral studies provides almost indisputable evidence for K+ channel dysfunction in the onset and maintenance of neuropathic pain. This idea is underlined by the observation that selective impairment of just one subtype of DRG K+ channel can produce signs of pain in vivo. Whilst it is established that various mediators, including cytokines and growth factors bring about injury-induced changes in DRG function and excitability, evidence presently available points to a seminal role for interleukin 1β (IL-1β) in control of K+ channel function. Despite the current state of knowledge, attempts to target K+ channels for therapeutic pain management have met with limited success. This situation may change with the advent of personalized medicine. Identification of specific sensory abnormalities and genetic profiling of individual patients may predict therapeutic benefit of K+ channel activators.
Collapse
Affiliation(s)
- Peter A. Smith
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
11
|
Korczeniewska OA, Khan J, Eliav E, Benoliel R. Molecular mechanisms of painful traumatic trigeminal neuropathy-Evidence from animal research and clinical correlates. J Oral Pathol Med 2020; 49:580-589. [PMID: 32557871 DOI: 10.1111/jop.13078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023]
Abstract
Painful traumatic trigeminal neuropathy (PTTN) may occur following major craniofacial or oral trauma, or may be subsequent to relatively minor dental interventions. Following injury, pain may originate from a peripheral nerve, a ganglion, or from the central nervous system. In this review, we focus on molecular mechanisms of pain resulting from injury to the peripheral branch of the trigeminal nerve. This syndrome has been termed painful traumatic trigeminal neuropathy (PTTN) by the International Headache Society and replaces previous terms including atypical odontalgia, deafferentation pain, traumatic neuropathy and phantom toothache. We emphasize the scientific evidence supporting the events purported to lead to PTTN by reviewing the pathophysiology of PTTN based on relevant animal models. Additionally, we briefly overview clinical correlates and pathophysiological manifestations of PTTN.
Collapse
Affiliation(s)
- Olga A Korczeniewska
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Junad Khan
- Eastman Institute of Oral Health, University of Rochester Medical Center, Rochester, New Jersey, USA
| | - Eli Eliav
- Eastman Institute of Oral Health, University of Rochester Medical Center, Rochester, New Jersey, USA
| | - Rafael Benoliel
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| |
Collapse
|
12
|
Decreased neural expression of the noradrenaline transporter in the papillary dermis after partial sciatic nerve lesion. J Chem Neuroanat 2020; 107:101806. [PMID: 32473320 DOI: 10.1016/j.jchemneu.2020.101806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 11/21/2022]
Abstract
After peripheral nerve injury, regeneration or collateral sprouting of noradrenergic nerve fibres in the papillary dermis of the injured limb may contribute to sympathetically-maintained pain. The aim of this study was to determine whether noradrenergic nerve fibre regeneration after partial sciatic nerve ligation (PSL) in Wistar rats was accompanied by parallel shifts in expression of the noradrenaline transporter (NAT). Four or 28 days after PSL surgery, immunohistochemistry was used to examine NAT expression in plantar hind paw skin in relation to pan-neuronal markers (class III beta-tubulin and protein gene product 9.5), peptidergic afferents containing calcitonin gene-related peptide (CGRP), nonpeptidergic afferents labelled by isolectin B4 (IB4), and tyrosine hydroxylase (TH), a marker for cutaneous noradrenergic nerve fibres. Most dermal nerve fibre populations decreased shortly after PSL. However, four weeks after PSL, an increase in staining intensity of CGRP and novel expression of TH were observed in the papillary dermis on the injured side. In contrast, neural expression of NAT was reduced in this region. Loss of NAT might have implications for sympathetically-maintained pain, as failure to rapidly clear noradrenaline could exacerbate aberrant sympathetic-sensory signalling between closely apposed noradrenergic and peptidergic nerve fibres.
Collapse
|
13
|
Leibovich H, Buzaglo N, Tsuriel S, Peretz L, Caspi Y, Katz B, Lev S, Lichtstein D, Binshtok AM. Abnormal Reinnervation of Denervated Areas Following Nerve Injury Facilitates Neuropathic Pain. Cells 2020; 9:cells9041007. [PMID: 32325693 PMCID: PMC7226396 DOI: 10.3390/cells9041007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 01/23/2023] Open
Abstract
An injury to peripheral nerves leads to skin denervation, which often is followed by increased pain sensitivity of the denervated areas and the development of neuropathic pain. Changes in innervation patterns during the reinnervation process of the denervated skin could contribute to the development of neuropathic pain. Here, we examined the changes in the innervation pattern during reinnervation and correlated them with the symptoms of neuropathic pain. Using a multispectral labeling technique—PainBow, which we developed, we characterized dorsal root ganglion (DRG) neurons innervating distinct areas of the rats’ paw. We then used spared nerve injury, causing partial denervation of the paw, and examined the changes in innervation patterns of the denervated areas during the development of allodynia and hyperalgesia. We found that, differently from normal conditions, during the development of neuropathic pain, these areas were mainly innervated by large, non-nociceptive neurons. Moreover, we found that the development of neuropathic pain is correlated with an overall decrease in the number of DRG neurons innervating these areas. Importantly, treatment with ouabain facilitated reinnervation and alleviated neuropathic pain. Our results suggest that local changes in peripheral innervation following denervation contribute to neuropathic pain development. The reversal of these changes decreases neuropathic pain.
Collapse
Affiliation(s)
- Hodaya Leibovich
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
| | - Nahum Buzaglo
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
| | - Shlomo Tsuriel
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
| | - Liat Peretz
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
| | - Yaki Caspi
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
| | - Ben Katz
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
| | - Shaya Lev
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
| | - David Lichtstein
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
| | - Alexander M. Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, Jerusalem 91120, Israel
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem 91120, Israel
- Correspondence: ; Tel.: +972-2-675-7349
| |
Collapse
|
14
|
Khan J, Zusman T, Wang Q, Eliav E. Acute and Chronic Pain in Orofacial Trauma Patients. J Endod 2019; 45:S28-S38. [DOI: 10.1016/j.joen.2019.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
15
|
Khan J, Zusman T, Wang Q, Eliav E. Acute and chronic pain in orofacial trauma patients. Dent Traumatol 2019; 35:348-357. [PMID: 31125489 DOI: 10.1111/edt.12493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 01/07/2023]
Abstract
Trauma or injury to the dentition and supporting tissues is associated with pain and discomfort, as expected, that may present immediately, shortly afterwards, or within a few days. Pain is an essential response to injury because it allows the organism to develop avoidance behavior to potential threats and helps the organism to avoid usage of the injured organ during the healing process. Not only does external trauma induce pain, but also essential invasive dental procedures such as extractions, dental implant insertions, root canal treatments, and oral surgeries are accompanied by similar post-surgical (post-traumatic) pain. The pain intensity after trauma varies and does not always correlate with the extent of injury. Trauma to the orofacial region or the teeth may also indirectly affect and induce pain in other orofacial structures such as the masticatory muscles, the temporomandibular joint, and even the cervical spine. In most cases, the pain will resolve as soon as healing of the affected tissue occurs or after dental and routine palliative treatment. In a limited number of cases, the pain persists beyond healing and evolves into a chronic pain state. Chronic pain in the orofacial region presents diagnostic and management challenges. Misdiagnosis or delayed diagnosis of the oral chronic pain condition may lead to unnecessary dental treatment. This article will discuss diagnosis and treatment for acute and chronic pain as well as potential mechanisms involved in the undesirable transition from acute to chronic pain.
Collapse
Affiliation(s)
- Junad Khan
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, Rochester, NY, USA
| | - Tal Zusman
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, Rochester, NY, USA
| | - Qian Wang
- Orofacial Pain and TMJ Disorders, Eastman Institute for Oral Health, Rochester, NY, USA
| | - Eli Eliav
- Eastman Institute for Oral Health, Rochester, NY, USA
| |
Collapse
|
16
|
Crawford LK, Caterina MJ. Functional Anatomy of the Sensory Nervous System: Updates From the Neuroscience Bench. Toxicol Pathol 2019; 48:174-189. [PMID: 31554486 DOI: 10.1177/0192623319869011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The simple tripartite classification of sensory neurons as A-beta, A-delta, and C fibers fails to convey the complexity of the neurons that encode stimuli as diverse as the texture of a surface, the location of a pinprick, or the direction of hair movement as a breeze moves across the skin. It has also proven to be inadequate when investigating the molecular mechanisms underlying pain, which can encompass any combination of chemical, tactile, and thermal modalities. Beginning with a brief overview of visceral and sensory neuroanatomy, this review expands upon sensory innervation of the skin as a prime example of the heterogeneity and complexity of the somatosensory nervous system. Neuroscientists have characterized defining features of over 15 subtypes of sensory neurons that innervate the skin of the mouse. This has enabled the study of cell-specific mechanisms of pain, which suggests that diverse sensory neuron subtypes may have distinct susceptibilities to toxic injury and different roles in pathologic mechanisms underlying altered sensation. Leveraging this growing body of knowledge for preclinical trials and models of neurotoxicity can vastly improve our understanding of peripheral nervous system dysfunction, advancing the fields of toxicologic pathology and neuropathology alike.
Collapse
Affiliation(s)
- LaTasha K Crawford
- Department of Pathobiological Sciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, USA, Madison, WI, USA
| | - Michael J Caterina
- Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Meng C, Yang X, Liu Y, Zhou Y, Rui J, Li S, Xu C, Zhuang Y, Lao J, Zhao X. Decreased expression of lncRNA Malat1 in rat spinal cord contributes to neuropathic pain by increasing neuron excitability after brachial plexus avulsion. J Pain Res 2019; 12:1297-1310. [PMID: 31114309 PMCID: PMC6497903 DOI: 10.2147/jpr.s195117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/15/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose: Neuropathic pain (NP) is a challenging clinical problem due to its complex pathogenesis. In our previous study using microarray, we found that the levels of lncRNA Malat1 were decreased in the spinal cord of NP rat after brachial plexus avulsion, but its contribution to NP remain unclear. The purpose of this study was to investigate its role in the pathogenesis of NP. Methods: In the NP model of complete brachial plexus avulsion rat, spinal cords were harvested, and fluorescence in situ hybridization (FISH) was used to test the spatial expression of Malat1 and qRT-PCR was used to confirm the quantitative expression of Malat1. In primary cultured neurons, Malat1 expression interfered with adenovirus. Spontaneous electric activities of neurons were tested using multi-electrode arrays and apoptosis of neurons was tested using TUNEL method. The change of intracellular calcium concentration was analyzed using calcium imaging method. Results: Decreased Malat1 expression was confirmed using qRT-PCR, and Malat1 was identified in the cytoplasm of neurons in spinal cord, but not in glia. In vitro, the decrease of Malat1 resulted in an increase in the frequency of spontaneous electric activity in neurons but had no effect on neuronal apoptosis. Further analysis indicated during glutamate stimulation, the change of intracellular calcium concentration in neurons with downregulated Malat1 expression was significantly greater than that in normal neurons. Conclusion: Reduced Malat1 expression may induce NP by increasing neuronal excitability in the spinal cord via regulation of calcium flux.
Collapse
Affiliation(s)
- Chong Meng
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Xun Yang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Yuzhou Liu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Yingjie Zhou
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Jing Rui
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Shenqian Li
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Ce Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Yongqing Zhuang
- Hand Surgery Department, Shenzhen People's Hospital, Shenzhen 518020, People's Republic of China
| | - Jie Lao
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| | - Xin Zhao
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,Key Laboratory of Hand Reconstruction, Ministry of Health, Shanghai 200032, People's Republic of China.,Shanghai Key Laboratory of Peripheral Nerve and Microsurgery, Shanghai 200032, People's Republic of China
| |
Collapse
|
18
|
Okada S, Saito H, Matsuura Y, Mikuzuki L, Sugawara S, Onose H, Asaka J, Ohara K, Lee J, Iinuma T, Katagiri A, Iwata K. Upregulation of calcitonin gene-related peptide, neuronal nitric oxide synthase, and phosphorylated extracellular signal-regulated kinase 1/2 in the trigeminal ganglion after bright light stimulation of the eye in rats. J Oral Sci 2019; 61:146-155. [PMID: 30918211 DOI: 10.2334/josnusd.18-0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Bright light stimulation of the eye activates trigeminal subnucleus caudalis (Vc) neurons in rats. Sensory information is conveyed to the Vc via the trigeminal ganglion (TG). Thus, it is likely that TG neurons respond to photic stimulation and are involved in photic hypersensitivity. However, the mechanisms underlying this process are unclear. Therefore, the hypothesis in this study is bright light stimulation enhances the excitability of TG neurons involved in photic hypersensitivity. Expressions of calcitonin gene-related peptide (CGRP) and neuronal nitric oxide synthase (nNOS) were significantly higher in TG neurons from 5 min to 12 h after photic stimulation of the eye. Phosphorylation of extracellular signal-regulated kinase1/2 (pERK1/2) was enhanced in TG neurons within 5 min after photic stimulation, while pERK1/2 immunoreactivity in satellite glial cells (SGCs) persisted for more than 12 h after the stimulus. Activation of SGCs was observed from 5 min to 2 h. Expression of CGRP, nNOS, and pERK1/2 was observed in small and medium TG neurons, and activation of SGCs and pERK1/2-immunoreactive SGCs encircling large TG neurons was accelerated after stimulation. These results suggest that upregulation of CGRP, nNOS, and pERK1/2 within the TG is involved in photic hypersensitivity.
Collapse
Affiliation(s)
- Shinji Okada
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry.,Department of Physiology, Nihon University School of Dentistry
| | - Hiroto Saito
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry.,Department of Physiology, Nihon University School of Dentistry
| | - Yutaka Matsuura
- Department of Oral Physiology, Osaka University Graduate School of Dentistry
| | - Lou Mikuzuki
- Department of Physiology, Nihon University School of Dentistry.,Department of Psychosomatic Dentistry, Tokyo Medical and Dental University, Graduate School
| | - Shiori Sugawara
- Department of Physiology, Nihon University School of Dentistry.,Department of Psychosomatic Dentistry, Tokyo Medical and Dental University, Graduate School
| | - Hiroki Onose
- Department of Physiology, Nihon University School of Dentistry
| | - Junichi Asaka
- Department of Physiology, Nihon University School of Dentistry
| | - Kinuyo Ohara
- Department of Endodontics, Nihon University School of Dentistry
| | - Jun Lee
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry
| | - Toshimitsu Iinuma
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry
| | - Ayano Katagiri
- Department of Physiology, Nihon University School of Dentistry.,Department of Oral Physiology, Osaka University Graduate School of Dentistry
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry
| |
Collapse
|
19
|
Phosphorylated CRMP2 Regulates Spinal Nociceptive Neurotransmission. Mol Neurobiol 2018; 56:5241-5255. [PMID: 30565051 DOI: 10.1007/s12035-018-1445-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/03/2018] [Indexed: 01/01/2023]
Abstract
The collapsin response mediator protein 2 (CRMP2) has emerged as a central node in assembling nociceptive signaling complexes involving voltage-gated ion channels. Concerted actions of post-translational modifications, phosphorylation and SUMOylation, of CRMP2 contribute to regulation of pathological pain states. In the present study, we demonstrate a novel role for CRMP2 in spinal nociceptive transmission. We found that, of six possible post-translational modifications, three phosphorylation sites on CRMP2 were critical for regulating calcium influx in dorsal root ganglion sensory neurons. Of these, only CRMP2 phosphorylated at serine 522 by cyclin-dependent kinase 5 (Cdk5) contributed to spinal neurotransmission in a bidirectional manner. Accordingly, expression of a non-phosphorylatable CRMP2 (S522A) decreased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs), whereas expression of a constitutively phosphorylated CRMP2 (S522D) increased the frequency of sEPSCs. The presynaptic nature of CRMP2's actions was further confirmed by pharmacological antagonism of Cdk5-mediated CRMP2 phosphorylation with S-N-benzy-2-acetamido-3-methoxypropionamide ((S)-lacosamide; (S)-LCM) which (i) decreased sEPSC frequency, (ii) increased paired-pulse ratio, and (iii) reduced the presynaptic distribution of CaV2.2 and NaV1.7, two voltage-gated ion channels implicated in nociceptive signaling. (S)-LCM also inhibited depolarization-evoked release of the pro-nociceptive neurotransmitter calcitonin gene-related peptide (CGRP) in the spinal cord. Increased CRMP2 phosphorylation in rats with spared nerve injury (SNI) was decreased by intrathecal administration of (S)-LCM resulting in a loss of presynaptic localization of CaV2.2 and NaV1.7. Together, these findings indicate that CRMP2 regulates presynaptic excitatory neurotransmission in spinal cord and may play an important role in regulating pathological pain. Novel targeting strategies to inhibit CRMP2 phosphorylation by Cdk5 may have great potential for the treatment of chronic pain.
Collapse
|
20
|
North RY, Lazaro TT, Dougherty PM. Ectopic Spontaneous Afferent Activity and Neuropathic Pain. Neurosurgery 2018; 65:49-54. [PMID: 31076785 DOI: 10.1093/neuros/nyy119] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Tyler T Lazaro
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Patrick M Dougherty
- The Departments of Pain Medicine Research, The Division of Anesthesia, Critical Care and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
21
|
The Neuroplastic and Therapeutic Potential of Spinal Interneurons in the Injured Spinal Cord. Trends Neurosci 2018; 41:625-639. [PMID: 30017476 DOI: 10.1016/j.tins.2018.06.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 12/25/2022]
Abstract
The central nervous system is not a static, hard-wired organ. Examples of neuroplasticity, whether at the level of the synapse, the cell, or within and between circuits, can be found during development, throughout the progression of disease, or after injury. One essential component of the molecular, anatomical, and functional changes associated with neuroplasticity is the spinal interneuron (SpIN). Here, we draw on recent multidisciplinary studies to identify and interrogate subsets of SpINs and their roles in locomotor and respiratory circuits. We highlight some of the recent progress that elucidates the importance of SpINs in circuits affected by spinal cord injury (SCI), especially those within respiratory networks; we also discuss potential ways that spinal neuroplasticity can be therapeutically harnessed for recovery.
Collapse
|
22
|
Zou Y, Xu F, Tang Z, Zhong T, Cao J, Guo Q, Huang C. Distinct calcitonin gene-related peptide expression pattern in primary afferents contribute to different neuropathic symptoms following chronic constriction or crush injuries to the rat sciatic nerve. Mol Pain 2018; 12:1744806916681566. [PMID: 28256957 PMCID: PMC5521344 DOI: 10.1177/1744806916681566] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Although calcitonin gene-related peptide is a recognized pain transducer, the expression of calcitonin gene-related peptide in primary afferents may be differentially affected following different types of nerve injury. Here, we examined whether different calcitonin gene-related peptide expression patterns in primary afferents contributes to distinct sensory disturbances in three animal models of sciatic nerve injury: chronic constriction injury, mild (100g force) or strong (1000g force) transient crush in rats. Assessments of withdrawal reflexes and spontaneous behavior indicated that chronic constriction injury and mild crush resulted in positive neuropathic symptoms (static/dynamic mechanical allodynia, heat hyperalgesia, cold allodynia, spontaneous pain). However, strong crush led to both positive (dynamic mechanical allodynia, cold allodynia, spontaneous pain) and negative symptoms (static mechanical hypoesthesia, heat hypoalgesia). Calcitonin gene-related peptide immunoreactivity in dorsal root ganglia and corresponding spinal cord segments, and calcitonin gene-related peptide mRNA levels in dorsal root ganglia, indicated that the primary afferent calcitonin gene-related peptide supply was markedly reduced only after strong crush. This reduction paralleled the development of negative symptoms (static mechanical hypoesthesia and heat hypoalgesia). Administration of exogenous calcitonin gene-related peptide intrathecally after strong crush did not alter heat hypoalgesia but ameliorated static mechanical hypoesthesia, an effect blocked by a calcitonin gene-related peptide receptor antagonist. Thus, reducing the primary afferent calcitonin gene-related peptide supply contributed to subsequent negative neuropathic symptoms, especially to static mechanical stimuli. Moreover, nerve injury caused a subcellular redistribution of calcitonin gene-related peptide from small- and medium-size dorsal root ganglia neurons to large-size dorsal root ganglia neurons, which paralleled the development of positive neuropathic symptoms. Intrathecal administration of the calcitonin gene-related peptide receptor antagonist ameliorated these positive symptoms, indicating that the expression of calcitonin gene-related peptide in large-size dorsal root ganglia neurons is important for the positive neuropathic symptoms in all three models. Taken together, these results suggest that distinct calcitonin gene-related peptide expression pattern in primary afferents contribute to different neuropathic symptoms following chronic constriction or crush injuries to the rat sciatic nerve.
Collapse
Affiliation(s)
- Yu Zou
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Fangting Xu
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Zhaohui Tang
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Tao Zhong
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Jiawei Cao
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Qulian Guo
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| | - Changsheng Huang
- 1 Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
23
|
Mikuzuki L, Saito H, Katagiri A, Okada S, Sugawara S, Kubo A, Ohara K, Lee J, Toyofuku A, Iwata K. Phenotypic change in trigeminal ganglion neurons associated with satellite cell activation via extracellular signal-regulated kinase phosphorylation is involved in lingual neuropathic pain. Eur J Neurosci 2017; 46:2190-2202. [PMID: 28834578 DOI: 10.1111/ejn.13667] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 12/22/2022]
Abstract
Iatrogenic trigeminal nerve injuries remain a common and complex clinical problem. Satellite glial cell (SGC) activation, associated phosphorylation of extracellular signal-regulated kinase (ERK), and neuropeptide expression in the trigeminal ganglion (TG) are known to be involved in trigeminal neuropathic pain related to trigeminal nerve injury. However, the involvement of these molecules in orofacial neuropathic pain mechanisms is still unknown. Phosphorylation of ERK1/2 in lingual nerve crush (LNC) rats was observed in SGCs. To evaluate the role of neuron-SGC interactions under neuropathic pain, calcitonin gene-related peptide (CGRP)-immunoreactive (IR), phosphorylated ERK1/2 (pERK1/2)-IR and glial fibrillary acidic protein (GFAP)-IR cells in the TG were studied in LNC rats. The number of CGRP-IR neurons and neurons encircled with pERK1/2-IR SGCs was significantly larger in LNC rats compared with sham rats. The percentage of large-sized CGRP-IR neurons was significantly higher in LNC rats. The number of CGRP-IR neurons, neurons encircled with pERK1/2-IR SGCs, and neurons encircled with GFAP-IR SGCs was decreased following CGRP receptor blocker CGRP8-37 or mitogen-activated protein kinase/ERK kinase 1 inhibitor PD98059 administration into the TG after LNC. Reduced thresholds to mechanical and heat stimulation to the tongue in LNC rats were also significantly recovered following CGRP8-37 or PD98059 administration. The present findings suggest that CGRP released from TG neurons activates SGCs through ERK1/2 phosphorylation and TG neuronal activity is enhanced, resulting in the tongue hypersensitivity associated with lingual nerve injury. The phenotypic switching of large myelinated TG neurons expressing CGRP may account for the pathogenesis of tongue neuropathic pain.
Collapse
Affiliation(s)
- Lou Mikuzuki
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Department of Psychosomatic Dentistry, Tokyo Medical and Dental University (TMDU) Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Hiroto Saito
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| | - Ayano Katagiri
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Shinji Okada
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| | - Shiori Sugawara
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Department of Psychosomatic Dentistry, Tokyo Medical and Dental University (TMDU) Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Asako Kubo
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kinuyo Ohara
- Department of Endodontics, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| | - Jun Lee
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| | - Akira Toyofuku
- Department of Psychosomatic Dentistry, Tokyo Medical and Dental University (TMDU) Graduate School, Bunkyo-ku, Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
24
|
Stubhaug A. Why are some patients with chronic pain from anterior abdominal nerve entrapment syndrome (ACNES) refractory to peripheral treatment with neurectomy? Scand J Pain 2017; 14:80-81. [PMID: 28850439 DOI: 10.1016/j.sjpain.2016.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Audun Stubhaug
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Gautier A, Geny D, Bourgoin S, Bernard J, Hamon M. Differential innervation of superficial versus deep laminae of the dorsal horn by bulbo-spinal serotonergic pathways in the rat. IBRO Rep 2017; 2:72-80. [PMID: 30135935 PMCID: PMC6084826 DOI: 10.1016/j.ibror.2017.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/28/2017] [Accepted: 04/07/2017] [Indexed: 01/04/2023] Open
Abstract
Convergent data showed that bulbo-spinal serotonergic projections exert complex modulatory influences on nociceptive signaling within the dorsal horn. These neurons are located in the B3 area which comprises the median raphe magnus (RMg) and the lateral paragigantocellular reticular (LPGi) nuclei. Because LPGi 5-HT neurons differ from RMg 5-HT neurons regarding both their respective electrophysiological properties and responses to noxious stimuli, we used anatomical approaches for further characterization of the respective spinal projections of LPGi versus RMg 5-HT neuron subgroups. Adult Sprague-Dawley rats were stereotaxically injected into the RMg or the LPGi with the anterograde tracer Phaseolus vulgaris leucoagglutinin (PHA-L). The precise location of injection sites and RMg vs LPGi spinal projections into the different dorsal horn laminae were visualized by PHA-L immunolabeling. Double immunofluorescent labeling of PHA-L and the serotonin transporter (5-HTT) allowed detection of serotonergic fibers among bulbo-spinal projections. Anterograde tracing showed that RMg neurons project preferentially into the deep laminae V-VI whereas LPGi neuron projections are confined to the superficial laminae I-II of the ipsilateral dorsal horn. All along the spinal cord, double-labeled PHA-L/5-HTT immunoreactive fibers, which represent only 5-15% of all PHA-L-immunoreactive projections, exhibit the same differential locations depending on their origin in the RMg versus the LPGi. The clear-cut distinction between dorsal horn laminae receiving bulbo-spinal serotonergic projections from the RMg versus the LPGi provides further anatomical support to the idea that the descending serotonergic pathways issued from these two bulbar nuclei might exert different modulatory influences on the spinal relay of pain signaling neuronal pathways.
Collapse
Affiliation(s)
| | | | | | | | - M. Hamon
- INSERM UMR 894 - Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| |
Collapse
|
26
|
Abstract
Definition and taxonomy This review deals with neuropathic pain of traumatic origin affecting the trigeminal nerve, i.e. painful post-traumatic trigeminal neuropathy (PTTN). Symptomatology The clinical characteristics of PTTN vary considerably, partly due to the type and extent of injury. Symptoms involve combinations of spontaneous and evoked pain and of positive and negative somatosensory signs. These patients are at risk of going through unnecessary dental/surgical procedures in the attempt to eradicate the cause of the pain, due to the fact that most dentists only rarely encounter PTTN. Epidemiology Overall, approximately 3% of patients with trigeminal nerve injuries develop PTTN. Patients are most often female above the age of 45 years, and both physical and psychological comorbidities are common. Pathophysiology PTTN shares many pathophysiological mechanisms with other peripheral neuropathic pain conditions. Diagnostic considerations PTTN may be confused with one of the regional neuralgias or other orofacial pain conditions. For intraoral PTTN, early stages are often misdiagnosed as odontogenic pain. Pain management Management of PTTN generally follows recommendations for peripheral neuropathic pain. Expert opinion International consensus on classification and taxonomy is urgently needed in order to advance the field related to this condition.
Collapse
Affiliation(s)
- Lene Baad-Hansen
- 1 Section of Orofacial Pain and Jaw Function, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark.,2 Scandinavian Center for Orofacial Neurosciences (SCON), Denmark/Sweden
| | - Rafael Benoliel
- 3 Rutgers School of Dental Medicine, Rutgers State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
27
|
Iyengar S, Ossipov MH, Johnson KW. The role of calcitonin gene-related peptide in peripheral and central pain mechanisms including migraine. Pain 2017; 158:543-559. [PMID: 28301400 PMCID: PMC5359791 DOI: 10.1097/j.pain.0000000000000831] [Citation(s) in RCA: 379] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/09/2016] [Accepted: 12/22/2016] [Indexed: 12/25/2022]
Abstract
Calcitonin gene-related peptide (CGRP) is a 37-amino acid peptide found primarily in the C and Aδ sensory fibers arising from the dorsal root and trigeminal ganglia, as well as the central nervous system. Calcitonin gene-related peptide was found to play important roles in cardiovascular, digestive, and sensory functions. Although the vasodilatory properties of CGRP are well documented, its somatosensory function regarding modulation of neuronal sensitization and of enhanced pain has received considerable attention recently. Growing evidence indicates that CGRP plays a key role in the development of peripheral sensitization and the associated enhanced pain. Calcitonin gene-related peptide is implicated in the development of neurogenic inflammation and it is upregulated in conditions of inflammatory and neuropathic pain. It is most likely that CGRP facilitates nociceptive transmission and contributes to the development and maintenance of a sensitized, hyperresponsive state not only of the primary afferent sensory neurons but also of the second-order pain transmission neurons within the central nervous system, thus contributing to central sensitization as well. The maintenance of a sensitized neuronal condition is believed to be an important factor underlying migraine. Recent successful clinical studies have shown that blocking the function of CGRP can alleviate migraine. However, the mechanisms through which CGRP may contribute to migraine are still not fully understood. We reviewed the role of CGRP in primary afferents, the dorsal root ganglion, and in the trigeminal system as well as its role in peripheral and central sensitization and its potential contribution to pain processing and to migraine.
Collapse
|
28
|
Benoliel R, Teich S, Eliav E. Painful Traumatic Trigeminal Neuropathy. Oral Maxillofac Surg Clin North Am 2016; 28:371-80. [DOI: 10.1016/j.coms.2016.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
29
|
Abstract
UNLABELLED The sensation of touch is initiated when fast conducting low-threshold mechanoreceptors (Aβ-LTMRs) generate impulses at their terminals in the skin. Plasticity in this system is evident in the process of adaption, in which a period of diminished sensitivity follows prior stimulation. CaMKII is an ideal candidate for mediating activity-dependent plasticity in touch because it shifts into an enhanced activation state after neuronal depolarizations and can thereby reflect past firing history. Here we show that sensory neuron CaMKII autophosphorylation encodes the level of Aβ-LTMR activity in rat models of sensory deprivation (whisker clipping, tail suspension, casting). Blockade of CaMKII signaling limits normal adaptation of action potential generation in Aβ-LTMRs in excised skin. CaMKII activity is also required for natural filtering of impulse trains as they travel through the sensory neuron T-junction in the DRG. Blockade of CaMKII selectively in presynaptic Aβ-LTMRs removes dorsal horn inhibition that otherwise prevents Aβ-LTMR input from activating nociceptive lamina I neurons. Together, these consequences of reduced CaMKII function in Aβ-LTMRs cause low-intensity mechanical stimulation to produce pain behavior. We conclude that, without normal sensory activity to maintain adequate levels of CaMKII function, the touch pathway shifts into a pain system. In the clinical setting, sensory disuse may be a critical factor that enhances and prolongs chronic pain initiated by other conditions. SIGNIFICANCE STATEMENT The sensation of touch is served by specialized sensory neurons termed low-threshold mechanoreceptors (LTMRs). We examined the role of CaMKII in regulating the function of these neurons. Loss of CaMKII function, such as occurred in rats during sensory deprivation, elevated the generation and propagation of impulses by LTMRs, and altered the spinal cord circuitry in such a way that low-threshold mechanical stimuli produced pain behavior. Because limbs are protected from use during a painful condition, this sensitization of LTMRs may perpetuate pain and prevent functional rehabilitation.
Collapse
|
30
|
Stemkowski PL, Noh MC, Chen Y, Smith PA. Increased excitability of medium-sized dorsal root ganglion neurons by prolonged interleukin-1β exposure is K(+) channel dependent and reversible. J Physiol 2015; 593:3739-55. [PMID: 26110238 PMCID: PMC4560594 DOI: 10.1113/jp270905] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/17/2015] [Indexed: 01/27/2023] Open
Abstract
KEY POINTS Neuropathic pain resulting from peripheral nerve injury is initiated and maintained by persistent ectopic activity in primary afferent neurons. Sciatic nerve injury increases the excitability of medium-sized dorsal root ganglion (DRG) neurons. Levels of the inflammatory cytokine interleukin 1β (IL-1β) increase and peak after 7 days. Five to six days of exposure of medium sized DRG neurons to 100 pm IL-1β promotes persistent increases in excitability which abate within 3-4 days of cytokine removal. This is associated with a profound attenuation of K(+) channel currents but only modest increases in function of cyclic nucleotide-sensitive hyperpolarization-activated channels (HCNs) and of voltage-gated Na(+) and Ca(2+) channel currents. It is unlikely, therefore, that direct interaction of IL-1β with DRG neurons is capable of initiating an enduring phenotypic shift in their electrophysiological properties that follows sciatic nerve injury. The findings also underline the importance of K(+) channel modulation in the actions of inflammatory mediators on peripheral neurons. ABSTRACT Chronic constriction injury of rat sciatic nerve promotes signs of neuropathic pain. This is associated with an increase in the level of interleukin 1β (IL-1β) in primary afferents that peaks at 7 days. This initial cytokine exposure has been proposed to trigger an enduring alteration in neuronal phenotype that underlies chronic hyper-excitability in sensory nerves, which initiates and maintains chronic neuropathic pain. We have shown previously that 5-6 days of exposure of rat dorsal root ganglia (DRGs) to 100 pm IL-1β increases the excitability of medium-sized neurons. We have now found using whole-cell recording that this increased excitability reverts to control levels within 3-4 days of cytokine removal. The effects of IL-1β were dominated by changes in K(+) currents. Thus, the amplitudes of A-current, delayed rectifier and Ca(2+) -sensitive K(+) currents were reduced by ∼68%, ∼64% and ∼36%, respectively. Effects of IL-1β on other cation currents were modest by comparison. There was thus a slight decrease in availability of high voltage-activated Ca(2+) channel current, a small increase in rates of activation of hyperpolarization-activated cyclic nucleotide-gated channel current (IH ), and a shift in the voltage dependence of activation of tetrodotoxin-sensitive sodium current (TTX-S INa ) to more negative potentials. It is unlikely, therefore, that direct interaction of IL-1β with DRG neurons initiates an enduring phenotypic shift in their electrophysiological properties following sciatic nerve injury. Persistent increases in primary afferent excitability following nerve injury may instead depend on altered K(+) channel function and on the continued presence of slightly elevated levels IL-1β and other cytokines.
Collapse
Affiliation(s)
- Patrick L Stemkowski
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada, T2N 4N1
| | - Myung-Chul Noh
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Yishen Chen
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Peter A Smith
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| |
Collapse
|
31
|
Effects of Vibration Therapy on Immobilization-Induced Hypersensitivity in Rats. Phys Ther 2015; 95:1015-26. [PMID: 25655883 DOI: 10.2522/ptj.20140137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 01/28/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cast immobilization induces mechanical hypersensitivity, which disturbs rehabilitation. Although vibration therapy can reduce various types of pain, whether vibration reduces immobilization-induced hypersensitivity remains unclear. OBJECTIVE The purpose of this study was to investigate the preventive and therapeutic effects of vibration therapy on immobilization-induced hypersensitivity. DESIGN The experimental design of the study involved conducting behavioral, histological, and immunohistochemical studies in model rats. METHODS Thirty-five Wistar rats (8 weeks old, all male) were used. The right ankle joints of 30 rats were immobilized by plaster cast for 8 weeks, and 5 rats were used as controls. The immobilized rats were divided randomly into the following 3 groups: (1) immobilization-only group (Im, n=10); (2) vibration therapy group 1, for which vibration therapy was initiated immediately after the onset of immobilization (Im+Vib1, n=10); and (3) vibration therapy group 2, for which vibration therapy was initiated 4 weeks after the onset of immobilization (Im+Vib2, n=10). Vibration was applied to the hind paw. The mechanical hypersensitivity and epidermal thickness of the hind paw skin were measured. To investigate central sensitization, calcitonin gene-related peptide (CGRP) expression in the spinal cord and dorsal root ganglion (DRG) was analyzed. RESULTS Immobilization-induced hypersensitivity was inhibited in the Im+Vib1 group but not in the Im+Vib2 group. Central sensitization, which was indicated by increases in CGRP expression in the spinal cord and the size of the area of CGRP-positive neurons in the DRG, was inhibited in only the Im+Vib1 group. Epidermal thickness was not affected by vibration stimulation. LIMITATIONS A limitation of this study is that the results were limited to an animal model and cannot be generalized to humans. CONCLUSIONS The data suggest that initiation of vibration therapy in the early phase of immobilization may inhibit the development of immobilization-induced hypersensitivity.
Collapse
|
32
|
Yin R, Liu D, Chhoa M, Li CM, Luo Y, Zhang M, Lehto SG, Immke DC, Moyer BD. Voltage-gated sodium channel function and expression in injured and uninjured rat dorsal root ganglia neurons. Int J Neurosci 2015; 126:182-92. [DOI: 10.3109/00207454.2015.1004172] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
33
|
Gilron I, Baron R, Jensen T. Neuropathic pain: principles of diagnosis and treatment. Mayo Clin Proc 2015; 90:532-45. [PMID: 25841257 DOI: 10.1016/j.mayocp.2015.01.018] [Citation(s) in RCA: 348] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/21/2015] [Accepted: 01/22/2015] [Indexed: 12/14/2022]
Abstract
Neuropathic pain is caused by disease or injury of the nervous system and includes various chronic conditions that, together, affect up to 8% of the population. A substantial body of neuropathic pain research points to several important contributory mechanisms including aberrant ectopic activity in nociceptive nerves, peripheral and central sensitization, impaired inhibitory modulation, and pathological activation of microglia. Clinical evaluation of neuropathic pain requires a thorough history and physical examination to identify characteristic signs and symptoms. In many cases, other laboratory investigations and clinical neurophysiological testing may help identify the underlying etiology and guide treatment selection. Available treatments essentially provide only symptomatic relief and may include nonpharmacological, pharmacological, and interventional therapies. Most extensive evidence is available for pharmacological treatment, and currently recommended first-line treatments include antidepressants (tricyclic agents and serotonin-norepinephrine reuptake inhibitors) and anticonvulsants (gabapentin and pregabalin). Individualized multidisciplinary patient care is facilitated by careful consideration of pain-related disability (eg, depression and occupational dysfunction) as well as patient education; repeat follow-up and strategic referral to appropriate medical/surgical subspecialties; and physical and psychological therapies. In the near future, continued preclinical and clinical research and development are expected to lead to further advancements in the diagnosis and treatment of neuropathic pain.
Collapse
Affiliation(s)
- Ian Gilron
- Departments of Anesthesiology and Perioperative Medicine and Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Troels Jensen
- Department of Neurology and Danish Pain Research Center, Aarhus University Hospital, Aarhus C, Denmark
| |
Collapse
|
34
|
Zhou C, Luo ZD. Nerve injury-induced calcium channel alpha-2-delta-1 protein dysregulation leads to increased pre-synaptic excitatory input into deep dorsal horn neurons and neuropathic allodynia. Eur J Pain 2015; 19:1267-76. [PMID: 25691360 DOI: 10.1002/ejp.656] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Up-regulation of voltage-gated calcium channel α2 δ1 subunit post spinal nerve ligation (SNL) injury or in α2 δ1 -overexpressing transgenic (Tg) mice correlates with tactile allodynia, a pain state mediated mainly by Aβ sensory fibres forming synaptic connections with deep dorsal horn (DDH) neurons. It is not clear, however, whether dysregulated α2 δ1 alters DDH synaptic neurotransmission that underlies tactile allodynia development post nerve injury. METHODS Tactile allodynia was tested in the SNL and α2 δ1 Tg models. Miniature excitatory/inhibitory post-synaptic currents were recorded in DDH neurons from these animal models using whole-cell patch clamp slice recording techniques. RESULTS There was a significant increase in the frequency, but not amplitude, of miniature excitatory post-synaptic currents (mEPSC) in DDH neurons that correlated with tactile allodynia in SNL and α2 δ1 Tg mice. Gabapentin, an α2 δ1 ligand that is known to block tactile allodynia in these models, also normalized mEPSC frequency dose-dependently in DDH neurons from SNL and α2 δ1 Tg mice. In contrast, neither frequency nor amplitude of miniature inhibitory post-synaptic currents was altered in DDH neurons from SNL and α2 δ1 Tg mice. CONCLUSION Our data suggest that α2 δ1 dysregulation is highly likely contributing to tactile allodynia through a pre-synaptic mechanism involving facilitation of excitatory synaptic neurotransmission in DDH of spinal cord.
Collapse
Affiliation(s)
- C Zhou
- Department of Pharmacology, School of Medicine, University of California Irvine, USA
| | - Z D Luo
- Department of Pharmacology, School of Medicine, University of California Irvine, USA.,Department of Anesthesiology and Perioperative Care, School of Medicine, University of California Irvine, USA
| |
Collapse
|
35
|
Neuropathies douloureuses et atteinte des petites fibres. Rev Neurol (Paris) 2014; 170:825-36. [DOI: 10.1016/j.neurol.2014.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 10/10/2014] [Indexed: 12/16/2022]
|
36
|
Fried K, Hansson P. The curse of a little of that human touch: Is CGRP responsible? Exp Neurol 2014; 255:83-5. [DOI: 10.1016/j.expneurol.2014.02.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/17/2014] [Accepted: 02/21/2014] [Indexed: 10/25/2022]
|
37
|
Bali KK, Hackenberg M, Lubin A, Kuner R, Devor M. Sources of individual variability: miRNAs that predispose to neuropathic pain identified using genome-wide sequencing. Mol Pain 2014; 10:22. [PMID: 24642266 PMCID: PMC4113183 DOI: 10.1186/1744-8069-10-22] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/26/2014] [Indexed: 01/19/2023] Open
Abstract
Background We carried out a genome-wide study, using microRNA sequencing (miRNA-seq), aimed at identifying miRNAs in primary sensory neurons that are associated with neuropathic pain. Such scans usually yield long lists of transcripts regulated by nerve injury, but not necessarily related to pain. To overcome this we tried a novel search strategy: identification of transcripts regulated differentially by nerve injury in rat lines very similar except for a contrasting pain phenotype. Dorsal root ganglia (DRGs) L4 and 5 in the two lines were excised 3 days after spinal nerve ligation surgery (SNL) and small RNAs were extracted and sequenced. Results We identified 284 mature miRNA species expressed in rat DRGs, including several not previously reported, and 3340 unique small RNA sequences. Baseline expression of miRNA was nearly identical in the two rat lines, consistent with their shared genetic background. In both lines many miRNAs were nominally up- or down-regulated following SNL, but the change was similar across lines. Only 3 miRNAs that were expressed abundantly (rno-miR-30d-5p, rno-miR-125b-5p) or at moderate levels (rno-miR-379-5p) were differentially regulated. This makes them prime candidates as novel PNS determinants of neuropathic pain. The first two are known miRNA regulators of the expression of Tnf, Bdnf and Stat3, gene products intimately associated with neuropathic pain phenotype. A few non-miRNA, small noncoding RNAs (sncRNAs) were also differentially regulated. Conclusions Despite its genome-wide coverage, our search strategy yielded a remarkably short list of neuropathic pain-related miRNAs. As 2 of the 3 are validated regulators of important pro-nociceptive compounds, it is likely that they contribute to the orchestration of gene expression changes that determine individual variability in pain phenotype. Further research is required to determine whether some of the other known or predicted gene targets of these miRNAs, or of the differentially regulated non-miRNA sncRNAs, also contribute.
Collapse
Affiliation(s)
| | | | | | | | - Marshall Devor
- Department of Cell & Developmental Biology, Institute of Life Sciences and Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| |
Collapse
|
38
|
Drummond PD, Drummond ES, Dawson LF, Mitchell V, Finch PM, Vaughan CW, Phillips JK. Upregulation of α1-adrenoceptors on cutaneous nerve fibres after partial sciatic nerve ligation and in complex regional pain syndrome type II. Pain 2014; 155:606-616. [DOI: 10.1016/j.pain.2013.12.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 12/07/2013] [Accepted: 12/10/2013] [Indexed: 10/25/2022]
|
39
|
Steinhoff MS, von Mentzer B, Geppetti P, Pothoulakis C, Bunnett NW. Tachykinins and their receptors: contributions to physiological control and the mechanisms of disease. Physiol Rev 2014; 94:265-301. [PMID: 24382888 DOI: 10.1152/physrev.00031.2013] [Citation(s) in RCA: 435] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The tachykinins, exemplified by substance P, are one of the most intensively studied neuropeptide families. They comprise a series of structurally related peptides that derive from alternate processing of three Tac genes and are expressed throughout the nervous and immune systems. Tachykinins interact with three neurokinin G protein-coupled receptors. The signaling, trafficking, and regulation of neurokinin receptors have also been topics of intense study. Tachykinins participate in important physiological processes in the nervous, immune, gastrointestinal, respiratory, urogenital, and dermal systems, including inflammation, nociception, smooth muscle contractility, epithelial secretion, and proliferation. They contribute to multiple diseases processes, including acute and chronic inflammation and pain, fibrosis, affective and addictive disorders, functional disorders of the intestine and urinary bladder, infection, and cancer. Neurokinin receptor antagonists are selective, potent, and show efficacy in models of disease. In clinical trials there is a singular success: neurokinin 1 receptor antagonists to treat nausea and vomiting. New information about the involvement of tachykinins in infection, fibrosis, and pruritus justifies further trials. A deeper understanding of disease mechanisms is required for the development of more predictive experimental models, and for the design and interpretation of clinical trials. Knowledge of neurokinin receptor structure, and the development of targeting strategies to disrupt disease-relevant subcellular signaling of neurokinin receptors, may refine the next generation of neurokinin receptor antagonists.
Collapse
|
40
|
Hamaue Y, Nakano J, Sekino Y, Chuganji S, Sakamoto J, Yoshimura T, Origuchi T, Okita M. Immobilization-induced hypersensitivity associated with spinal cord sensitization during cast immobilization and after cast removal in rats. J Physiol Sci 2013; 63:401-8. [PMID: 23818166 PMCID: PMC10717811 DOI: 10.1007/s12576-013-0277-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/14/2013] [Indexed: 01/10/2023]
Abstract
This study examined mechanical and thermal hypersensitivity in the rat hind paw during cast immobilization of the hind limbs for 4 or 8 weeks and following cast removal. Blood flow, skin temperature, and volume of the rat hind paw were assessed in order to determine peripheral circulation of the hind limbs. Sensitization was analyzed by measuring the expression of the calcitonin gene-related peptide (CGRP) in the spinal dorsal horn following cast immobilization. Two weeks post immobilization, mechanical and thermal sensitivities increased significantly in all rats; however, peripheral circulation was not affected by immobilization. Cast immobilization for 8 weeks induced more serious hypersensitivity compared to cast immobilization for 4 weeks. Moreover, CGRP expression in the deeper lamina layer of the spinal dorsal horn increased in the rats immobilized for 8 weeks but not in those immobilized for 4 weeks. These findings suggest that immobilization-induced hypersensitivity develops during the immobilization period without affecting peripheral circulation. Our results also highlight the possibility that prolonged immobilization induces central sensitization in the spinal cord.
Collapse
Affiliation(s)
- Yohei Hamaue
- Department of Locomotive Rehabilitation Science, Unit of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
- Department of Rehabilitation, Juzenkai Hospital, 7-18 Kago-machi, Nagasaki, 850-0905 Japan
| | - Jiro Nakano
- Unit of Physical and Occupational Therapy Sciences, Nagasaki University Graduate School of Biochemical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
| | - Yuki Sekino
- Department of Locomotive Rehabilitation Science, Unit of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
| | - Sayaka Chuganji
- Unit of Physical and Occupational Therapy Sciences, Nagasaki University Graduate School of Biochemical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
| | - Jyunya Sakamoto
- Department of Rehabilitation, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Toshiro Yoshimura
- Department of Locomotive Rehabilitation Science, Unit of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
| | - Tomoki Origuchi
- Department of Locomotive Rehabilitation Science, Unit of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
| | - Minoru Okita
- Department of Locomotive Rehabilitation Science, Unit of Rehabilitation Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8520 Japan
| |
Collapse
|
41
|
Dynamic genotype-selective "phenotypic switching" of CGRP expression contributes to differential neuropathic pain phenotype. Exp Neurol 2013; 250:194-204. [PMID: 24076003 DOI: 10.1016/j.expneurol.2013.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 08/18/2013] [Accepted: 09/16/2013] [Indexed: 11/23/2022]
Abstract
Using a genetic model we demonstrate the role played by "phenotypic switching" of calcitonin gene related peptide (CGRP) expression in axotomized large Aβ afferents in the development of neuropathic pain behavior in rats. After nerve injury both substance P and CGRP are upregulated in Aβ afferents in the corresponding DRGs. It has been proposed that intraspinal release of these neurotransmitters upon gentle stroking of skin drives ascending pain signaling pathways resulting in tactile allodynia. We reported previously that in rat lines genetically selected for high (HA) vs. low (LA) pain phenotype, SP is upregulated equally in both strains, but that CGRP is upregulated exclusively in the pain prone HA line (Nitzan-Luques et al., 2011). This implicates CGRP as the principal driver of tactile allodynia. Here we confirm this conclusion by showing: 1) that the time of emergence of CGRP-IR in DRG Aβ neurons and their central terminals in HA rats matches that of pain behavior, 2) that following spinal nerve lesion (SNL) selective activation of low threshold afferents indeed drives postsynaptic pain-signaling neurons and induces central sensitization in HA rats, as monitored using c-Fos as a marker. These changes are much less prominent in LA rats, 3) that intrathecal (i.t.) administration of CGRP induces tactile allodynia in naïve rats and 4) that i.t. administration of the CGRP-receptor antagonist BIBN4096BS (Olcegepant) attenuates SNL-evoked tactile allodynia, without blocking baseline nociception. Together, these observations support the hypothesis that genotype-selective phenotypic switching of CGRP expression in Aβ afferents following nerve injury is a fundamental mechanism of neuropathic tactile allodynia.
Collapse
|
42
|
Szajkowski S, Marcol W, Właszczuk A, Cieślar G, Pietrucha-Dutczak M, Sieroń A, Lewin-Kowalik J. The influence of spatial pulsed magnetic field application on neuropathic pain after tibial nerve transection in rat. Electromagn Biol Med 2013; 33:35-46. [PMID: 23781991 DOI: 10.3109/15368378.2013.783849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The purpose of the study was to examine the influence of the spatial variable magnetic field (induction: 150-300 µT, 80-150 µT, 20-80 µT; frequency 40 Hz) on neuropathic pain after tibial nerve transection. The experiments were carried out on 64 male Wistar C rats. The exposure of animals to magnetic field was performed 1 d/20 min., 5 d/week, for 28 d. Behavioural tests assessing the intensity of allodynia and sensitivity to mechanical and thermal stimuli were conducted 1 d prior to surgery and 3, 7, 14, 21 and 28 d after the surgery. The extent of autotomy was examined. Histological and immunohistochemical analysis was performed. The use of extremely low-frequency magnetic fields of minimal induction values (20-80 µT/40 Hz) decreased pain in rats after nerve transection. The nociceptive sensitivity of healthy rats was not changed following the exposition to the spatial magnetic field of the low frequency. The results of histological and immunohistochemical investigations confirm those findings. Our results indicate that extremely low-frequency magnetic field may be useful in the neuropathic pain therapy.
Collapse
|
43
|
Pitcher GM, Ritchie J, Henry JL. Peripheral neuropathy induces cutaneous hypersensitivity in chronically spinalized rats. PAIN MEDICINE 2013; 14:1057-71. [PMID: 23855791 DOI: 10.1111/pme.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES The present study was aimed at the issue of whether peripheral nerve injury-induced chronic pain is maintained by supraspinal structures governing descending facilitation to the spinal dorsal horn, or whether altered peripheral nociceptive mechanisms sustain central hyperexcitability and, in turn, neuropathic pain. We examined this question by determining the contribution of peripheral/spinal mechanisms, isolated from supraspinal influence(s), in cutaneous hypersensitivity in an animal model of peripheral neuropathy. METHODS Adult rats were spinalized at T8-T9; 8 days later, peripheral neuropathy was induced by implanting a 2-mm polyethylene cuff around the left sciatic nerve. Hind paw withdrawal responses to mechanical or thermal plantar stimulation were evaluated using von Frey filaments or a heat lamp, respectively. RESULTS Spinalized rats without cuff implantation exhibited a moderate decrease in mechanical withdrawal threshold on ~day 10 (P < 0.05) and in thermal withdrawal threshold on ~day 18 (P < 0.05). However, cuff-implanted spinalized rats developed a more rapid and significant decrease in mechanical (~day 4; P < 0.001) and thermal (~day 10; P < 0.05) withdrawal thresholds that remained significantly decreased through the duration of the study. CONCLUSIONS Our findings demonstrate an aberrant peripheral/spinal mechanism that induces and maintains thermal and to a greater degree tactile cutaneous hypersensitivity in the cuff model of neuropathic pain, and raise the prospect that altered peripheral/spinal nociceptive mechanisms in humans with peripheral neuropathy may have a pathologically relevant role in both inducing and sustaining neuropathic pain.
Collapse
Affiliation(s)
- Graham M Pitcher
- Departments of Physiology and Psychiatry, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
44
|
Long-term changes in trigeminal ganglionic and thalamic neuronal activities following inferior alveolar nerve transection in behaving rats. J Neurosci 2013; 32:16051-63. [PMID: 23136441 DOI: 10.1523/jneurosci.1828-12.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The transection of the inferior alveolar nerve (IANx) produces allodynia in the whisker pad (V2 division) of rats. Ectopic discharges from injured trigeminal ganglion (TG) neurons and thalamocortical reorganization are possible contributors to the sensitization of uninjured V2 primary and CNS neurons. To test which factor is more important, TG and ventroposterior medial nucleus (VPM) neurons were longitudinally followed before, during, and after IANx for up to 80 d. Spontaneous discharges and mechanical stimulation-evoked responses were recorded in conscious and in anesthetized states. Results show (1) a sequential increase in spontaneous activities, first in the injured TG neurons of the IAN (2-30 d), followed by uninjured V2 ganglion neurons (6-30 d), and then VPM V2 neurons (7-30 d) after IANx; (2) ectopic discharges included burst and regular firing patterns in the IAN and V2 branches of the TG neurons; and (3) the receptive field expanded, the modality shifted, and long-lasting after-discharges occurred only in VPM V2 neurons. All of these changes appeared in the late or maintenance phase (7-30 d) and disappeared during the recovery phase (40-60 d). These observations suggest that ectopic barrages in the injured IAN contribute more to the development of sensitization, whereas the modality shift and evoked after-discharges in the VPM thalamic neurons contribute more to the maintenance phase of allodynia by redirecting tactile information to the cortex as nociceptive.
Collapse
|
45
|
Gui Q, Xu C, Zhuang L, Xia S, Chen Y, Peng P, Yu S. A new rat model of bone cancer pain produced by rat breast cancer cells implantation of the shaft of femur at the third trochanter level. Cancer Biol Ther 2012; 14:193-9. [PMID: 23254954 DOI: 10.4161/cbt.23291] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Bone cancer pain remains one of the most challenging cancer pains to fully control. In order to clarify bone cancer pain mechanisms and examine treatments, animal models mimicking the human condition are required. In our model of Walker 256 tumor cells implantation of the shaft of femur at the third trochanter level, the anatomical structure is relatively simple and the drilled hole is vertical and in the cortical bone only 1-2 mm in depth without injury of the distal femur. Pain behaviors and tumor growth were observed for 21 days. And neurochemical changes were further investigated in this model. The results showed that cancer-bearing rats demonstrated a decreased limb use score from day 14, an increased spontaneous flinching and guarding times from day 7 and a decreased withdrawal threshold from day 6. The tumor infiltration of bone was monitored by MRI and further verified by histological examination. C-fos and the capsaicin receptor (TRPV1) positive neurons were more expressed in cancer-bearing rats and the substance P expression has no difference, suggesting that neurons were activated in the model. Our animal model demonstrated time-dependent tumor growth and pain behaviors and will be a novel animal model of bone cancer pain in the future.
Collapse
Affiliation(s)
- Qi Gui
- Department of Oncology, Tongji Hospital; Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
46
|
The effects of neuregulin-1β on neuronal phenotypes of primary cultured dorsal root ganglion neurons by activation of PI3K/Akt. Neurosci Lett 2012; 511:52-7. [DOI: 10.1016/j.neulet.2012.01.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 01/16/2012] [Accepted: 01/18/2012] [Indexed: 11/19/2022]
|
47
|
von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 2012; 73:638-52. [PMID: 22365541 PMCID: PMC3319438 DOI: 10.1016/j.neuron.2012.02.008] [Citation(s) in RCA: 593] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2012] [Indexed: 01/01/2023]
Abstract
After nerve injury maladaptive changes can occur in injured sensory neurons and along the entire nociceptive pathway within the CNS, which may lead to spontaneous pain or pain hypersensitivity. The resulting neuropathic pain syndromes present as a complex combination of negative and positive symptoms, which vary enormously from individual to individual. This variation depends on a diversity of underlying pathophysiological changes resulting from the convergence of etiological, genotypic, and environmental factors. The pain phenotype can serve therefore, as a window on underlying pathophysiological neural mechanisms and as a guide for developing personalized pain medicine.
Collapse
Affiliation(s)
- Christian A von Hehn
- FM Kirby Neurobiology Center, Children's Hospital Boston, and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|