1
|
Ikehata Y, Oshima E, Hayashi Y, Tanaka Y, Sato H, Hitomi S, Shiratori-Hayashi M, Urata K, Kimura Y, Shibuta I, Ohba S, Iwata K, Mizuta K, Shirota T, Shinoda M. Fibroblast-derived IL-33 exacerbates orofacial neuropathic pain via the activation of TRPA1 in trigeminal ganglion neurons. Brain Behav Immun 2025; 123:982-996. [PMID: 39500418 DOI: 10.1016/j.bbi.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/02/2024] [Accepted: 11/02/2024] [Indexed: 11/11/2024] Open
Abstract
Damage to the peripheral nerves of trigeminal ganglion (TG) neurons leads to intractable orofacial neuropathic pain through the induction of neuroinflammation. However, the details of this process are not yet fully understood. Here, we found that fibroblast-derived interleukin (IL)-33 was required for the development of mechanical allodynia in whisker pad skin following infraorbital nerve injury (IONI). The amount of IL-33 in the TG increased after IONI when the mice exhibited mechanical allodynia. Neutralization of IL-33 in the TG inhibited the development of IONI-induced mechanical allodynia. Conversely, intra-TG administration of recombinant human IL-33 (rhIL-33) elicited mechanical allodynia in naïve mice. IL-33 and its receptor were exclusively expressed in fibroblasts and neurons, respectively, in the TG. Fibroblast ablation caused the loss of IL-33 in the TG and delayed the development of mechanical allodynia after IONI. rhIL-33 elicited an increase in intracellular Ca2+ concentration and subsequent enhancement of Ca2+ influx via transient receptor potential ankyrin 1 (TRPA1) in primary cultured TG neurons. Additionally, rhIL-33 facilitated membrane translocation of TRPA1 in the TG. Mechanical allodynia caused by intra-TG administration of rhIL-33 was significantly inhibited by pharmacological blockade or gene silencing of TRPA1 in the TG. Inhibition of protein kinase A abrogated TRPA1 membrane translocation and delayed mechanical allodynia after IONI. Substance P stimulation caused upregulation of IL-33 expression in primary cultured fibroblasts. Preemptive administration of a neurokinin-1 receptor antagonist in the TG attenuated mechanical allodynia and IL-33 expression following IONI. Taken together, these results indicate that fibroblast-derived IL-33 exacerbates TG neuronal excitability via suppression of tumorigenicity 2 (ST2)-TRPA1 signaling, ultimately leading to orofacial neuropathic pain.
Collapse
Affiliation(s)
- Yousuke Ikehata
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 142-8515, Japan; Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Eri Oshima
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 142-8515, Japan; Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan.
| | - Yukinori Tanaka
- Division of Dento-oral Anesthesiology, Tohoku University Graduate School of Dentistry, Seiryomachi 4-1, Aoba-ku, Sendai 980-8575, Japan
| | - Hitoshi Sato
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 142-8515, Japan
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Miho Shiratori-Hayashi
- Department of Molecular and Systems Pharmacology, Faculty of Pharmacy, Juntendo University, 6-8-1, Hinode, Urayasu, Chiba 279-0013, Japan; Juntendo Itch Research Center, Institute for Environmental and Gender-Specific Medicine, Graduate School of Medicine, Juntendo University, 2-1-1, Tomioka, Urayasu, Chiba 279-0021, Japan
| | - Kentaro Urata
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Yuki Kimura
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Ikuko Shibuta
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Seigo Ohba
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 142-8515, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Kentaro Mizuta
- Division of Dento-oral Anesthesiology, Tohoku University Graduate School of Dentistry, Seiryomachi 4-1, Aoba-ku, Sendai 980-8575, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 142-8515, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13, Kandasurugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| |
Collapse
|
2
|
Niemeyer CS, Harlander-Locke M, Bubak AN, Rzasa-Lynn R, Birlea M. Trigeminal Postherpetic Neuralgia: From Pathophysiology to Treatment. Curr Pain Headache Rep 2024; 28:295-306. [PMID: 38261232 PMCID: PMC10940365 DOI: 10.1007/s11916-023-01209-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2023] [Indexed: 01/24/2024]
Abstract
PURPOSE OF REVIEW Trigeminal postherpetic neuralgia (TG-PHN) is a neuropathic pain condition complicating herpes zoster (HZ) attributed to the trigeminal nerve. It poses significant challenges due to its persistent and debilitating nature. This review explores the clinical characteristics of TG-PHN, analyzes its pathophysiological underpinnings, and addresses existent and potential therapies. RECENT FINDINGS TG-PHN is one of the most common and complex PHN locations. It has distinguishing clinical and pathophysiological characteristics, starting with viral triggered injuries to the trigeminal ganglion (TG) and peripheral tissue and involving the ascending and descending brain modulation pathways. Current therapies include vaccines, oral and topical medications, and interventional approaches, like nerve blocks and neurostimulation. This review covers TG-PHN's clinical and physiological components, treatment options, and potential future targets for improved management. By exploring the complexities of this condition, we aim to contribute to developing more effective and targeted therapies for patients suffering from trigeminal PHN.
Collapse
Affiliation(s)
- Christy S Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Harlander-Locke
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew N Bubak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rachael Rzasa-Lynn
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marius Birlea
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
3
|
Nakaya A, Kaneko K, Miyazawa K, Matsumoto A, Hisanaga K, Matsumori Y, Nagano I. Neuralgia in the occipital region associated with ipsilateral trigeminal herpes zoster: Three case reports. Headache 2024; 64:464-468. [PMID: 38525807 DOI: 10.1111/head.14698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Nerve fibers related to pain and temperature sensation in the trigeminal nerve territory converge with the upper cervical spinal nerves from the level of the lower medulla oblongata to the upper cervical cord. This structure is called the trigemino-cervical complex and may cause referred pain in the territory of the trigeminal or upper cervical spinal nerves. CASE SERIES Here, we report three cases of paroxysmal neuralgia in the occipital region with mild conjunctivitis or a few reddish spots in the ipsilateral trigeminal nerve territory. The patients exhibited gradual progression of these reddish spots evolving into vesicles over the course of several days, despite the absence of a rash in the occipital region. The patients were diagnosed with trigeminal herpes zoster and subsequently received antiherpetic therapy. Remarkably, the neuralgia in the occipital region showed gradual amelioration or complete resolution before the treatment, with no sequelae reported in the occipital region. DISCUSSION The trigemino-cervical complex has the potential to cause neuralgia in the occipital region, as referred pain, caused by trigeminal herpes zoster. These cases suggest that, even if conjunctivitis or reddish spots appear to be trivial in the trigeminal nerve territory, trigeminal herpes zoster should be considered when neuralgia occurs in the ipsilateral occipital region.
Collapse
Affiliation(s)
- Akihiko Nakaya
- Department of Neurology, National Hospital Organization Miyagi National Hospital, Yamamoto-cho, Miyagi, Japan
| | - Kimihiko Kaneko
- Department of Neurology, National Hospital Organization Miyagi National Hospital, Yamamoto-cho, Miyagi, Japan
| | - Koichi Miyazawa
- Department of Neurology, National Hospital Organization Miyagi National Hospital, Yamamoto-cho, Miyagi, Japan
- Department of Neurology, Tohoku Medical and Pharmaceutical University Hospital, Sendai, Miyagi, Japan
| | - Arifumi Matsumoto
- Department of Neurology, National Hospital Organization Miyagi National Hospital, Yamamoto-cho, Miyagi, Japan
| | - Kinya Hisanaga
- Department of Neurology, National Hospital Organization Miyagi National Hospital, Yamamoto-cho, Miyagi, Japan
| | | | - Isao Nagano
- Department of Neurology, National Hospital Organization Miyagi National Hospital, Yamamoto-cho, Miyagi, Japan
| |
Collapse
|
4
|
Lu J, Yang B, Zhang W, Cheng H, Zeng J, Wang Y, Wei W, Liu Z. Transplantation of olfactory ensheathing cells can alleviate neuroinflammatory responses in rats with trigeminal neuralgia. Brain Res 2024; 1825:148732. [PMID: 38104922 DOI: 10.1016/j.brainres.2023.148732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/07/2023] [Accepted: 12/14/2023] [Indexed: 12/19/2023]
Abstract
Trigeminal neuralgia (TN) is a common form of facial pain, which primarily manifests as severe pain similar to facial acupuncture and electric shock. Olfactory ensheathing cells (OECs) are glial cells with high bioactivity; these cells are essential for the periodic regeneration of the olfactory nerve and have been utilized for the repair of nerve injuries. A member of the P2X receptor family, P2X7R, is an ion channel type receptor that has been confirmed to participate in various pain response processes. In this study, we transplanted OECs into trigeminal nerve-model rats with distal infraorbital nerve ligation to observe the therapeutic effect of transplanted OECs in rats. Additionally, we utilized the P2X7R-specific inhibitor brilliant blue G (BBG) to study the therapeutic mechanisms of cell transplantation. The facial mechanical pain threshold of these rats significantly increased following cell transplantation. The immunohistochemistry, immunoblotting, and RT-qPCR results demonstrated that the levels of P2X7R, (NOD)-like receptor protein-3 (NLRP3), nuclear factor-κB (NF-κB), interleukin (IL)-1β, and IL-18 in the trigeminal ganglion of rats treated with OEC transplantation or BBG treatment were significantly lower than those in the injured group without treatment. Overall, our results demonstrate that OEC transplantation can alleviate TN in rats, and it can reduce the expression of P2X7R related inflammatory factors in TN rats, reducing neuroinflammatory response in TG.
Collapse
Affiliation(s)
- Jiafeng Lu
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Baolin Yang
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Wenjun Zhang
- Rehabilitation Medicine Department, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hui Cheng
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Jingnan Zeng
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Yuanli Wang
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Wei Wei
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Zengxu Liu
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
5
|
Zhang Y, Sun D, Xie Y, Li R, Zhao H, Wang Z, Feng L. Predictive value of preoperative magnetic resonance imaging structural and diffusion indices for the results of trigeminal neuralgia microvascular decompression surgery. Neuroradiology 2023:10.1007/s00234-023-03155-4. [PMID: 37140598 DOI: 10.1007/s00234-023-03155-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
PURPOSE To explore the predictive value of preoperative magnetic resonance imaging structural and diffusion indices of the spinal trigeminal tract (SpTV) on the results of microvascular decompression (MVD) in patients with trigeminal neuralgia (TN). METHODS This retrospective study included patients diagnosed with TN and treated with MVD in the Jining First People's Hospital between January 2020 and January 2021. The patients were divided into good and poor results groups according to postoperative pain relief. Logistic regression analysis was performed to explore independent risk factors for poor results of MVD, and their predictive value was examined using receiver operating characteristic (ROC) curves. RESULTS A total of 97 TN cases were included, 24 cases with a poor result and 73 with a good result. They were comparable in demographic characteristics. Fractional anisotropy (FA) was lower (P < 0.001), and radial diffusivity (RD) was higher (P < 0.001) in the poor result group compared to the good result group. Patients in the good result group showed a higher proportion of grade 3 neurovascular contact (NVC) (39.7% vs. 16.7%, P = 0.001) and a lower RD (P < 0.001). The multivariate analysis showed that the RD of SpTV (OR = 0.000016, 95% CI: 0.000-0.004, P < 0.001) and NVC (OR = 8.07, 95% CI: 1.67-38.93, P = 0.009) were independently associated with poor results. The area under the curve (AUC) of RD and NVC were 0.848 and 0.710, and their combination achieved an AUC of 0.880. CONCLUSION NVC and RD of SpTV are independent risk factors for poor results after MVD surgery, and combining the NVC and RD might achieve relatively high predictive value for poor results.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China
| | - Dengbin Sun
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China
| | - Yunjie Xie
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China
| | - Rui Li
- Department of Radiology, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China
| | - Hang Zhao
- Department of Radiology, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China
| | - Zhaoping Wang
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China
| | - Lei Feng
- Department of Neurosurgery, Jining No. 1 People's Hospital, Jining, Shandong, 272001, People's Republic of China.
| |
Collapse
|
6
|
Basedau H, Oppermann T, Gundelwein Silva E, Peng KP, May A. Characterization of trigeminal C-fiber reactivity through capsaicin-induced release of calcitonin gene-related peptide. Headache 2023; 63:353-359. [PMID: 36705344 DOI: 10.1111/head.14471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/24/2022] [Accepted: 12/16/2022] [Indexed: 01/28/2023]
Abstract
OBJECTIVE We hypothesized that the response of trigeminal dermal blood flow (DBF) in the trigeminal system and consecutive expansion of flare response to capsaicin would differ from the somatosensory system (arm). We also investigated whether there are differences between patients with migraine and healthy controls (HC). BACKGROUND Functional differences between the trigeminal and extracephalic somatosensory systems may partly explain the susceptibility for headaches in patients with migraine. Capsaicin-induced activation of nociceptive C-fibers in the skin is mainly mediated by calcitonin gene-related peptide (CGRP) and induces cutaneous vessel dilatation and flare response. METHODS Female patients with migraine (n = 38) and age-matched HC (n = 35) underwent DBF measurement at baseline and after topical capsaicin administration using laser speckle imaging. DBF before and after capsaicin stimulation was analyzed over ophthalmic nerve/maxillary nerve/mandibular nerve (V1/V2/V3) dermatomes and the forearm as an extracephalic control. RESULTS Capsaicin-induced DBF increased more in the trigeminal dermatomes than on the forearm. The V1 dermatome showed a smaller increase of DBF in patients with migraine compared to HC. CONCLUSION Our results suggest that the trigeminovascular system reacts differently from extracephalic areas, which may explain the trigeminal susceptibility to CGRP-mediated pain attacks. By demonstrating a different reactivity of the V1 dermatome in patients with migraine, our finding suggests that the first trigeminal branch is functionally different from the second and third branches; however, only in patients with migraine.
Collapse
Affiliation(s)
- Hauke Basedau
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf Martinistr, Hamburg, Germany
| | - Thalea Oppermann
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf Martinistr, Hamburg, Germany
| | - Elisa Gundelwein Silva
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf Martinistr, Hamburg, Germany
| | - Kuan-Po Peng
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf Martinistr, Hamburg, Germany
| | - Arne May
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf Martinistr, Hamburg, Germany
| |
Collapse
|
7
|
Hornung RS, Kinchington PR, Umorin M, Kramer PR. PAQR8 and PAQR9 expression is altered in the ventral tegmental area of aged rats infected with varicella zoster virus. Mol Pain 2023; 19:17448069231202598. [PMID: 37699860 PMCID: PMC10515525 DOI: 10.1177/17448069231202598] [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: 07/06/2023] [Revised: 08/17/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Infection with varicella zoster virus (VZV) results in chicken pox and reactivation of VZV results in herpes zoster (HZ) or what is often referred to as shingles. Patients with HZ experience decreased motivation and increased emotional distress consistent with functions of the ventral tegmental area (VTA) of the brain. In addition, activity within the ventral tegmental area is altered in patients with HZ. HZ primarily affects individuals that are older and the VTA changes with age. To begin to determine if the VTA has a role in HZ symptoms, a screen of 10,000 genes within the VTA in young and old male rats was completed after injecting the whisker pad with VZV. The two genes that had maximal change were membrane progesterone receptors PAQR8 (mPRβ) and PAQR9 (mPRε). Neurons and non-neuronal cells expressed both PAQR8 and PAQR9. PAQR8 and PAQR9 protein expression was significantly reduced after VZV injection of young males. In old rats PAQR9 protein expression was significantly increased after VZV injection and PAQR9 protein expression was reduced in aged male rats versus young rats. Consistent with previous results, pain significantly increased after VZV injection of the whisker pad and aged animals showed significantly more pain than young animals. Our data suggests that PAQR8 and PAQR9 expression is altered by VZV injection and that these changes are affected by age.
Collapse
Affiliation(s)
- Rebecca S Hornung
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Paul R Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mikhail Umorin
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Phillip R Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| |
Collapse
|
8
|
Abstract
Trigeminal neuralgia (TN) is a rare neuropathic pain disorder characterized by recurrent, paroxysmal episodes of short-lasting severe electric shock-like pain along the sensory distribution of the trigeminal nerve. Recent classification systems group TN into 3 main categories depending on the underlying pathophysiology. This article will present a case history and review the epidemiology, diagnostic criteria, classification, clinical features, diagnostic investigations, pathophysiology, and management of TN.
Collapse
Affiliation(s)
- Shehryar Nasir Khawaja
- Orofacial Pain Medicine, Department of Internal Medicine, Shaukat Khanum Memorial Cancer Hospital & Research Centers, Lahore, Pakistan.
| | - Steven J Scrivani
- Department of Diagnostic Sciences, Craniofacial Pain Center, Tufts University School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Kc E, Islam J, Park YS. Trigeminal ganglion itself can be a viable target to manage trigeminal neuralgia. J Headache Pain 2022; 23:150. [PMID: 36424545 PMCID: PMC9686102 DOI: 10.1186/s10194-022-01512-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022] Open
Abstract
Excruciating trigeminal neuralgia (TN) management is very difficult and severely affects the patient's quality of life. Earlier studies have shown that the trigeminal ganglion (TG) comprises several receptors and signal molecules that are involved in the process of peripheral sensitization, which influences the development and persistence of neuropathic pain. Targeting TG can modulate this sensitization pathway and mediate the pain-relieving effect. So far,there are few studies in which modulation approaches to TG itself have been suggested so far. "Trigeminal ganglion modulation" and "trigeminal neuralgia" were used as search phrases in the Scopus Index and PubMed databases to discover articles that were pertinent to the topic. In this review, we address the role of the trigeminal ganglion in TN and underlying molecules and neuropeptides implicated in trigeminal pain pathways in processing pathological orofacial pain. We also reviewed different modulation approaches in TG for TN management. Furthermore, we discuss the prospect of targeting trigeminal ganglion to manage such intractable pain.
Collapse
Affiliation(s)
- Elina Kc
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Jaisan Islam
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Young Seok Park
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, Korea.
| |
Collapse
|
10
|
Kobayashi S, O'Hashi K, Kobayashi M. Repetitive nociceptive stimulation increases spontaneous neural activation similar to nociception-induced activity in mouse insular cortex. Sci Rep 2022; 12:15190. [PMID: 36071208 PMCID: PMC9452502 DOI: 10.1038/s41598-022-19562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
Recent noninvasive neuroimaging technology has revealed that spatiotemporal patterns of cortical spontaneous activity observed in chronic pain patients are different from those in healthy subjects, suggesting that the spontaneous cortical activity plays a key role in the induction and/or maintenance of chronic pain. However, the mechanisms of the spontaneously emerging activities supposed to be induced by nociceptive inputs remain to be established. In the present study, we investigated spontaneous cortical activities in sessions before and after electrical stimulation of the periodontal ligament (PDL) by applying wide-field and two-photon calcium imaging to anesthetized GCaMP6s transgenic mice. First, we identified the sequential cortical activation patterns from the primary somatosensory and secondary somatosensory cortices to the insular cortex (IC) by PDL stimulation. We, then found that spontaneous IC activities that exhibited a similar spatiotemporal cortical pattern to evoked activities by PDL stimulation increased in the session after repetitive PDL stimulation. At the single-cell level, repetitive PDL stimulation augmented the synchronous neuronal activity. These results suggest that cortical plasticity induced by the repetitive stimulation leads to the frequent PDL stimulation-evoked-like spontaneous IC activation. This nociception-induced spontaneous activity in IC may be a part of mechanisms that induces chronic pain.
Collapse
Affiliation(s)
- Shutaro Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Department of Oral Surgery, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kazunori O'Hashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Molecular Imaging Research Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
11
|
Peng KP, Jürgens T, Basedau H, Ortlieb L, May A. Sumatriptan prevents central sensitisation specifically in the trigeminal dermatome in humans. Eur J Pain 2022; 26:2152-2161. [PMID: 36001070 DOI: 10.1002/ejp.2027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/22/2022] [Accepted: 08/20/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND The exact mechanism and site of action of triptans in aborting migraine attacks remain under debate. We hypothesized that the clinical efficacy of triptans lies in aborting central sensitization and focused on the question of why triptans are headache-specific, i.e. highly effective in migraine and cluster headache and ineffective in extracephalic pain. METHODS Forty healthy participants were enrolled in this double-blinded, randomized, placebo-controlled study. The effect of sumatriptan (n=20) vs placebo (n=20) was investigated in a cephalic (V1) vs an extracephalic dermatome (forearm) using a topical capsaicin sensitization model. Capsaicin-induced primary and secondary hyperalgesia were evaluated using quantitative sensory testing. RESULTS After capsaicin application, primary hyperalgesia developed in both sumatriptan and placebo groups in both dermatomes. However, sumatriptan exclusively prevented secondary hyperalgesia in the V1 dermatome but not on the forearm. Placebo exerted no effects on secondary hyperalgesia in both trigeminal and extracephalic dermatomes. Additionally, sumatriptan reduced the flare size exclusively in the V1 dermatome. CONCLUSIONS Our data suggest that sumatriptan reduces central sensitization (secondary hyperalgesia) without modulating peripheral sensitization (primary hyperalgesia) in a human pain model of capsaicin-induced sensitization. Moreover, despite a systemic administration of sumatriptan, the modulatory effects are trigeminal-specific, echoing the clinical effect of triptans in aborting headaches, but not extracephalic pain.
Collapse
Affiliation(s)
- Kuan-Po Peng
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Jürgens
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Neurology, KMG Medical Center, Güstrow, Germany
| | - Hauke Basedau
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Luise Ortlieb
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arne May
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Bayat AH, Azimi H, Hassani Moghaddam M, Ebrahimi V, Fathi M, Vakili K, Mahmoudiasl GR, Forouzesh M, Boroujeni ME, Nariman Z, Abbaszadeh HA, Aryan A, Aliaghaei A, Abdollahifar MA. COVID-19 causes neuronal degeneration and reduces neurogenesis in human hippocampus. Apoptosis 2022; 27:852-868. [PMID: 35876935 PMCID: PMC9310365 DOI: 10.1007/s10495-022-01754-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 11/30/2022]
Abstract
Recent investigations of COVID-19 have largely focused on the effects of this novel virus on the vital organs in order to efficiently assist individuals who have recovered from the disease. In the present study we used hippocampal tissue samples extracted from people who died after COVID-19. Utilizing histological techniques to analyze glial and neuronal cells we illuminated a massive degeneration of neuronal cells and changes in glial cells morphology in hippocampal samples. The results showed that in hippocampus of the studied brains there were morphological changes in pyramidal cells, an increase in apoptosis, a drop in neurogenesis, and change in spatial distribution of neurons in the pyramidal and granular layer. It was also demonstrated that COVID-19 alter the morphological characteristics and distribution of astrocyte and microglia cells. While the exact mechanism(s) by which the virus causes neuronal loss and morphology in the central nervous system (CNS) remains to be determined, it is necessary to monitor the effect of SARS-CoV-2 infection on CNS compartments like the hippocampus in future investigations. As a result of what happened in the hippocampus secondary to COVID-19, memory impairment may be a long-term neurological complication which can be a predisposing factor for neurodegenerative disorders through neuroinflammation and oxidative stress mechanisms.
Collapse
Affiliation(s)
- Amir-Hossein Bayat
- Department of Basic Sciences, Saveh University of Medical Sciences, Saveh, Iran
| | - Helia Azimi
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mahdi Forouzesh
- Legal Medicine Research Center, Iranian Legal Medicine Organization, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Zahra Nariman
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat-Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arefeh Aryan
- Anatomy Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Aliaghaei
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Amin Abdollahifar
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Department of Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Kramer PR, Umorin M, Hornung R, Benson MD, Kinchington PR. Sex Differences in the Role of Neurexin 3α in Zoster Associated Pain. Front Integr Neurosci 2022; 16:915797. [PMID: 35875508 PMCID: PMC9302461 DOI: 10.3389/fnint.2022.915797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Varicella zoster virus (VZV) induces orofacial pain and female rats show greater pain than male rats. During the proestrus phase of the estrous cycle the VZV induce pain response is attenuated in female rats. A screen of gene expression changes in diestrus and proestrus female rats indicated neurexin 3α (Nrxn3α) was elevated in the central amygdala of proestrus rats vs. diestrus rats. GABAergic neurons descend from the central amygdala to the lateral parabrachial region and Nrxn3α is important for presynaptic γ-Aminobutyric acid (GABA) release. Thus, we hypothesized that the reduced orofacial pain in male rats and proestrus female rats is the result of increased Nrxn3α within the central amygdala that increases GABA release from axon terminals within the parabrachial and inhibits ascending pain signals. To test this hypothesis Nrxn3 α expression was knocked-down by infusing shRNA constructs in the central amygdala. Then GABA release in the parabrachial was quantitated concomitant with measuring the pain response. Results revealed that knockdown of Nrxn3α expression significantly increases the pain response in both male rats and proestrus female rats vs. diestrus rats. GABA release was significantly reduced in the parabrachial of male and proestrus female rats after Nrxn3α knockdown. Neuronal activity of excitatory neurons was significantly inhibited in the parabrachial after Nrxn3α knockdown. These results are consistent with the idea that Nrxn3 within the central amygdala controls VZV associated pain by regulating GABA release in the lateral parabrachial that then modulates ascending orofacial pain signals.
Collapse
Affiliation(s)
- Phillip R. Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - Mikhail Umorin
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - Rebecca Hornung
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - M. Douglas Benson
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, United States
| | - Paul R. Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Neurexin 3α in the central amygdala has a role orofacial varicella zoster pain. Neuroscience 2022; 496:16-26. [PMID: 35679996 PMCID: PMC9329223 DOI: 10.1016/j.neuroscience.2022.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
Abstract
Varicella zoster virus (VZV) is responsible for chronic pain. VZV injection has similarities to herpes zoster (HZ) "shingles" pain in humans. In this study orofacial pain was induced by injecting male rats with the human VZV. The amygdala and parabrachial have been implicated to control affective/motivational orofacial pain. Recently our lab reported neurexin 3α (Nrxn3α) is expressed in the central amygdala and parabrachial. GABAergic neurons descend from the central amygdala to the lateral parabrachial region and Nrxn3α is important for presynaptic (γ-Aminobutyric acid) GABA release. Thus, we hypothesized that lateral parabrachial neuronal activity and orofacial pain are controlled by Nrxn3α within the central amygdala. To test the hypothesis Nrxn3α expression was knocked down (i.e., using short hairpin RNA or shRNA) in the central amygdala and GABA release and neuronal activity were quantitated in the parabrachial concomitant with measurement of the VZV induced pain response. Results revealed that attenuating Nrxn3 expression within the amygdala reduces GABA release in the parabrachial and increases neuronal activity within the lateral parabrachial region. Attenuating Nrxn3 expression also increases VZV associated orofacial pain. Activating GABAergic neurons within the central amygdala with opsins increase GABA release in the parabrachial and reduced the pain response after Nrxn3 shRNA treatment. These results are consistent with the idea that Nrxn3 within the central amygdala controls VZV associated pain by regulating GABA release in the lateral parabrachial that then controls the activity of ascending pain neurons.
Collapse
|
15
|
Trigeminal sensory modulatory effects of galcanezumab and clinical response prediction. Pain 2022; 163:2194-2199. [PMID: 35170575 DOI: 10.1097/j.pain.0000000000002614] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/09/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Galcanezumab, a monoclonal antibody against calcitonin gene-related peptide, is an emerging migraine preventative. We hypothesized that the preventive effects are conveyed via modulation of somatosensory processing and that certain sensory profiles may hence be associated with different clinical responses. We recruited migraine patients (n=26), who underwent quantitative sensory tests (QST) over the right V1 dermatome and forearm at baseline (T0), 2-3 weeks (T1), and one year (T12) after monthly galcanezumab treatment. The clinical response was defined as a reduction of ≥30% in headache frequency based on the headache diary. Predictors for clinical response were calculated using binary logistical regression models. After galcanezumab (T1 vs. T0), the heat pain threshold (HPT) (°C, 44.9 ± 3.4 vs. 43.0 ± 3.3, p=0.013) and mechanical pain threshold (MPT) (log mN, 1.60 ± 0.31 vs. 1.45 ± 0.26, p=0.042) were increased exclusively in the V1 dermatome, but not the forearm. These changes were immediate, did not differ between responders and non-responders, and did not last in one year of follow-up (T12 vs. T0). However, baseline HPT (OR: 2.13, 95% CI: 1.08-4.19, p = 0.029) on the forearm was a robust predictor for a clinical response three months later. In summary, our data demonstrated that galcanezumab modulates pain thresholds specifically in the V1 dermatome, but this modulation is short-lasting and irrelevant to clinical response. Instead, the clinical response may be determined by individual sensibility even before the administration of medication.
Collapse
|
16
|
Mulpuri Y, Yamamoto T, Nishimura I, Spigelman I. Role of voltage-gated sodium channels in axonal signal propagation of trigeminal ganglion neurons after infraorbital nerve entrapment. NEUROBIOLOGY OF PAIN 2022; 11:100084. [PMID: 35128176 PMCID: PMC8803652 DOI: 10.1016/j.ynpai.2022.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 11/25/2022]
Abstract
Infraorbital nerve entrapment (IoNE) induces mechanical allodynia and enhances signal propagation in primary afferent A- and C-fibers. IoNE increases sensitivity of A- and C-fibers to conduction block by tetrodotoxin (TTX) and selective voltage-gated sodium channel 1.8 (NaV1.8) inhibitor, A-803467. IoNE increases signal propagation in vibrissal pad Ad -, but not Aβ-fibers, and their sensitivity to conduction block by the selective NaV1.8 inhibitor. IoNE increases membrane excitability of dissociated small and medium sized trigeminal neurons. IoNE increases nerve, but not ganglion, levels of NaV1.3, NaV1.7, and NaV1.8 mRNAs, and NaV1.8 protein.
Chronic pain arising from peripheral nerve injuries represents a significant clinical challenge because even the most efficacious anticonvulsant drug treatments are limited by their side effects profile. We investigated pain behavior, changes in axonal signal conduction and excitability of trigeminal neurons, and expression of voltage-gated sodium channels (NaVs) in the infraorbital nerve and trigeminal ganglion (TG) after infraorbital nerve entrapment (IoNE). Compared to Sham, IoNE rats had increased A- and C-fiber compound action potentials (CAPs) and Aδ component of A-CAP area from fibers innervating the vibrissal pad. After IoNE, A- and C-fiber CAPs were more sensitive to blockade by tetrodotoxin (TTX), and those fibers that were TTX-resistant were more sensitive to blockade by the NaV1.8 selective blocker, A-803467. Although NaV1.7 blocker, ICA-121431 alone, did not affect Aδ-fiber signal propagation, cumulative application with A-803467 and 4,9-anhydro-TTX significantly reduced the Aδ-fiber CAP in IoNE rats. In patch clamp recordings from small- and medium-sized TG neurons, IoNE resulted in reduced action potential (AP) depolarizing current threshold, hyperpolarized AP voltage threshold, increased AP duration, and a more depolarized membrane potential. While the transcripts of most NaVs were reduced in the ipsilateral TG after IoNE, NaV1.3, NaV1.7, and NaV1.8 mRNAs, and NaV1.8 protein, were significantly increased in the nerve. Altogether, our data suggest that axonal redistribution of NaV1.8, and to a lesser extent NaV1.3, and NaV1.7 contributes to enhanced nociceptive signal propagation in peripheral nerve after IoNE.
Collapse
|
17
|
Zaman S, Kane T, Katta M, Georgiou M, Michaelides M. Photoaversion in inherited retinal diseases: clinical phenotypes, biological basis, and qualitative and quantitative assessment. Ophthalmic Genet 2021; 43:143-151. [PMID: 34957896 DOI: 10.1080/13816810.2021.2015789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Severe light sensitivity is a feature common to a range of ophthalmological and neurological diseases. In inherited retinal diseases (IRDs) particularly, this may be accompanied by significant visual disruption. These symptoms are extremely debilitating for affected individuals and have significant implications in terms of day-to-day activities. Underlying mechanisms remain to be fully elucidated. Currently, there are many assessments of photoaversion (PA), however, all have limitations, with quantitative measurement in particular needing further evaluation. To understand the complexities associated with photoaversion from different pathologies, qualitative and quantitative assessments of the light aversion response must be standardized. There is no treatment to date, and strategies to alleviate symptoms focus on light avoidance. With respect to IRDs, however, gene therapy is currently being investigated in clinical trials and promising and further treatments may be on the horizon. The better characterization of these symptoms is an important end point measure in IRD gene therapy trials.
Collapse
Affiliation(s)
- Serena Zaman
- Moorfields Eye Hospital, London, UK.,UCL Institute of Ophthalmology, University College London, London, UK
| | - Thomas Kane
- Moorfields Eye Hospital, London, UK.,UCL Institute of Ophthalmology, University College London, London, UK
| | - Mohamed Katta
- Moorfields Eye Hospital, London, UK.,UCL Institute of Ophthalmology, University College London, London, UK
| | - Michalis Georgiou
- Moorfields Eye Hospital, London, UK.,UCL Institute of Ophthalmology, University College London, London, UK
| | - Michel Michaelides
- Moorfields Eye Hospital, London, UK.,UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
18
|
Jafari Khaljiri H, Jamalkhah M, Amini Harandi A, Pakdaman H, Moradi M, Mowla A. Comprehensive Review on Neuro-COVID-19 Pathophysiology and Clinical Consequences. Neurotox Res 2021; 39:1613-1629. [PMID: 34169404 PMCID: PMC8225460 DOI: 10.1007/s12640-021-00389-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/08/2023]
Abstract
Aside from the respiratory distress as the predominant clinical presentation of SARS-CoV-2 infection, various neurological complications have been reported with the infection during the ongoing pandemic, some of which cause serious morbidity and mortality. Herein, we gather the latest anatomical evidence of the virus's presence within the central nervous system. We then delve into the possible SARS-CoV-2 entry routes into the neurological tissues, with the hematogenous and the neuronal routes as the two utmost passage routes into the nervous system. We then give a comprehensive review of the neurological manifestations of the SARS-CoV-2 invasion in both the central and peripheral nervous system and its underlying pathophysiology via investigating large studies in the field and case reports in cases of study scarcity.
Collapse
Affiliation(s)
- Helia Jafari Khaljiri
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Monire Jamalkhah
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Ali Amini Harandi
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hossein Pakdaman
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Moradi
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Mowla
- Division of Endovascular Neurosurgery, Department of Neurological Surgery, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA
| |
Collapse
|
19
|
Xu W, Yao Y, Zhu D, Han L, Wang L, Wang Y. Involvement of the BNP/NPR-A/BKCa pathway in rat trigeminal ganglia following chronic constriction injury. J Neurophysiol 2021; 125:1139-1145. [PMID: 33596737 DOI: 10.1152/jn.00682.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Accumulating evidence indicates that the brain natriuretic peptide (BNP) and its receptor (natriuretic peptide receptor, NPR) are widely distributed in a variety of tissues including trigeminal ganglion (TG). Furthermore, recent studies support the involvement of the BNP-NPR-A pathway in acute and chronic pain. To investigate the role of this pathway in chronic pain, an infraorbital nerve-chronic constriction injury (ION-CCI) model of trigeminal neuralgia (TN) was produced in the rat. The time course of changes in mechanical pain threshold was examined. We observed an upregulation of BNP and NPR-A and a downregulation of large-conductance Ca2+-activated K+ (BKCa) mRNA and protein in rats subjected to ION-CCI. Patch clamping experiments in vitro found that BKCa currents were significantly reduced in rats subjected to ION-CCI. BNP increased BKCa currents in ION-CCI rats. These results suggest that BNP and NPR-A might serve as endogenous pain relievers in ION-CCI rats. Modulation of the BNP/NPR-A/BKCa channel pathway in TG may be a viable strategy for the treatment of TN.NEW & NOTEWORTHY BNP has been known to activate its receptor, NPR-A, to modulate inflammatory pain. However, the potential modulatory roles of BNP in TN have not been investigated in detail. We established an ION-CCI model of TN in the rat and observed an upregulation of BNP and NPR-A and a downregulation of BKCa in rats subjected to ION-CCI. Moreover, BNP can increase BKCa currents in ION-CCI rats. Thus, BNP and NPR-A might have inhibitory effects on trigeminal neuralgia through activating the BNP/NPR-A/BKCa channel pathway.
Collapse
Affiliation(s)
- Wenhua Xu
- Key Lab of Oral Diseases Research of Anhui Province, Stomatologic Hospital & College, Anhui Medical University, Hefei, People's Republic of China
| | - Yuzhi Yao
- Key Lab of Oral Diseases Research of Anhui Province, Stomatologic Hospital & College, Anhui Medical University, Hefei, People's Republic of China
| | - Dawei Zhu
- Key Lab of Oral Diseases Research of Anhui Province, Stomatologic Hospital & College, Anhui Medical University, Hefei, People's Republic of China
| | - Liang Han
- Key Lab of Oral Diseases Research of Anhui Province, Stomatologic Hospital & College, Anhui Medical University, Hefei, People's Republic of China
| | - Liecheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| | - Yuanyin Wang
- Key Lab of Oral Diseases Research of Anhui Province, Stomatologic Hospital & College, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
20
|
Koh W, Lim H, Chen X. Atypical triggers in trigeminal neuralgia: the role of A-delta sensory afferents in food and weather triggers. Korean J Pain 2021; 34:66-71. [PMID: 33380569 PMCID: PMC7783845 DOI: 10.3344/kjp.2021.34.1.66] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 11/05/2022] Open
Abstract
Background Trigeminal neuralgia is a debilitating craniofacial pain syndrome that is characterized by paroxysms of intense, short-lived electric shock-like pains in the trigeminal nerve distribution. Recently, the presence of triggers has become one of the key diagnostic criteria in the 3rd edition of the International Classification of Headache Disorders. Light touch is the most common trigger, however other non-mechanical triggers, such as cold weather and certain foods, have been thought to provoke trigeminal neuralgia anecdotally. We aimed to characterize the prevalence and characteristics of these atypical triggers. Methods We conducted a retrospective, cross-sectional study of atypical triggers in trigeminal neuralgia patients seen in a tertiary pain clinic in Singapore. Patients were recruited via clinic records, and study data were identified from physician documentation. Results A total of 60 patients met the inclusion criteria. Weather triggers were observed in 12 patients (20%), of which five patients (8%) reported strong winds, 4 patients (7%) reported cold temperatures, and 3 patients (5%) reported cold winds as triggers. Fifteen patients (25%) had a specific food trigger, of which 10 patients (17%) reported hard or tough food, 5 patients (8%) reported hot/cold food, 4 patients (7%) reported spicy food, and 2 patients (3%) reported sweet food as triggers. Conclusions Although trigeminal neuralgia is most commonly triggered by mechanical stimuli, atypical triggers such as cold temperatures and certain foods are seen in a significant proportion of patients. These atypical triggers may share a common pathway of sensory afferent Aδ fiber activation.
Collapse
Affiliation(s)
- Wenjun Koh
- Department of Anaesthesiology, Singapore General Hospital, Singapore
| | - Huili Lim
- Department of Anaesthesiology, Singapore General Hospital, Singapore
| | - Xuanxuan Chen
- Department of Anaesthesiology, Singapore General Hospital, Singapore
| |
Collapse
|
21
|
Shih-Ping Hung P, Tohyama S, Zhang JY, Hodaie M. Temporal disconnection between pain relief and trigeminal nerve microstructural changes after Gamma Knife radiosurgery for trigeminal neuralgia. J Neurosurg 2020; 133:727-735. [PMID: 31299654 DOI: 10.3171/2019.4.jns19380] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/12/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Gamma Knife radiosurgery (GKRS) is a noninvasive surgical treatment option for patients with medically refractive classic trigeminal neuralgia (TN). The long-term microstructural consequences of radiosurgery and their association with pain relief remain unclear. To better understand this topic, the authors used diffusion tensor imaging (DTI) to characterize the effects of GKRS on trigeminal nerve microstructure over multiple posttreatment time points. METHODS Ninety-two sets of 3-T anatomical and diffusion-weighted MR images from 55 patients with TN treated by GKRS were divided within 6-, 12-, and 24-month posttreatment time points into responder and nonresponder subgroups (≥ 75% and < 75% reduction in posttreatment pain intensity, respectively). Within each subgroup, posttreatment pain intensity was then assessed against pretreatment levels and followed by DTI metric analyses, contrasting treated and contralateral control nerves to identify specific biomarkers of successful pain relief. RESULTS GKRS resulted in successful pain relief that was accompanied by asynchronous reductions in fractional anisotropy (FA), which maximized 24 months after treatment. While GKRS responders demonstrated significantly reduced FA within the radiosurgery target 12 and 24 months posttreatment (p < 0.05 and p < 0.01, respectively), nonresponders had statistically indistinguishable DTI metrics between nerve types at each time point. CONCLUSIONS Ultimately, this study serves as the first step toward an improved understanding of the long-term microstructural effect of radiosurgery on TN. Given that FA reductions remained specific to responders and were absent in nonresponders up to 24 months posttreatment, FA changes have the potential of serving as temporally consistent biomarkers of optimal pain relief following radiosurgical treatment for classic TN.
Collapse
Affiliation(s)
- Peter Shih-Ping Hung
- 1Division of Brain, Imaging & Behaviour-Systems Neuroscience, Krembil Research Institute, and
- 2Institute of Medical Science and
| | - Sarasa Tohyama
- 1Division of Brain, Imaging & Behaviour-Systems Neuroscience, Krembil Research Institute, and
- 2Institute of Medical Science and
| | - Jia Y Zhang
- 1Division of Brain, Imaging & Behaviour-Systems Neuroscience, Krembil Research Institute, and
| | - Mojgan Hodaie
- 1Division of Brain, Imaging & Behaviour-Systems Neuroscience, Krembil Research Institute, and
- 2Institute of Medical Science and
- 3Department of Surgery, Faculty of Medicine, University of Toronto, Ontario, Canada
- 4Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network; and
| |
Collapse
|
22
|
Hornung R, Pritchard A, Kinchington PR, Kramer PR. Reduced activity of GAD67 expressing cells in the reticular thalamus enhance thalamic excitatory activity and varicella zoster virus associated pain. Neurosci Lett 2020; 736:135287. [PMID: 32763361 DOI: 10.1016/j.neulet.2020.135287] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/23/2020] [Accepted: 07/30/2020] [Indexed: 11/16/2022]
Abstract
Within the reticular thalamic nucleus neurons express gamma aminobutyric acid (GABA) and these cells project to the ventral posteromedial thalamic nucleus. When GABA activity decreases the activity of excitatory cells in the ventral posteromedial nucleus would be expected to increase. In this study, we addressed the hypothesis that attenuating GABAergic cells in the reticular thalamic nucleus increases excitatory activity in the ventral posteromedial nucleus increasing varicella zoster virus (VZV) associated pain in the orofacial region. Adeno-associated virus (AAV) was infused in the reticular thalamic nucleus of Gad1-Cre rats. This virus transduced a G inhibitory designer receptor exclusively activated by designer drugs (DREADD) gene that was Cre dependent. A dose of estradiol that was previously shown to reduce VZV pain and increase GABAergic activity was administered to castrated and ovariectomized rats. Previous studies suggest that estradiol attenuates herpes zoster pain by increasing the activity of inhibitory neurons and decreasing the activity of excitatory cells within the lateral thalamic region. The ventral posteromedial nucleus was infused with AAV containing a GCaMP6f expression construct. A glass lens was implanted for miniscope imaging. Our results show that the activity of GABA cells within the reticular thalamic region decreased with clozapine N-oxide treatment concomitant with increased calcium activity of excitatory cells in the ventral posteromedial nucleus and an increased orofacial pain response. The results suggest that estradiol attenuates herpes zoster pain by increasing the activity of inhibitory neurons within the reticular thalamus that then inhibit excitatory activity in ventral posteromedial nucleus causing a reduction in orofacial pain.
Collapse
Affiliation(s)
- Rebecca Hornung
- Texas A&M University College of Dentistry, Dallas, TX, 75246, United States
| | - Addison Pritchard
- Texas A&M University College of Dentistry, Dallas, TX, 75246, United States
| | - Paul R Kinchington
- Dept Ophthalmology, Molecular Genetics and Biochemistry, The Campbell Laboratory for Infectious Eye Diseases, University of Pittsburgh School of Medicine, University of Pittsburg, 203 Lothrop St., Pittsburgh, PA, 15213, United States
| | - Phillip R Kramer
- Texas A&M University College of Dentistry, Dallas, TX, 75246, United States.
| |
Collapse
|
23
|
DosSantos MF, Devalle S, Aran V, Capra D, Roque NR, Coelho-Aguiar JDM, Spohr TCLDSE, Subilhaga JG, Pereira CM, D'Andrea Meira I, Niemeyer Soares Filho P, Moura-Neto V. Neuromechanisms of SARS-CoV-2: A Review. Front Neuroanat 2020; 14:37. [PMID: 32612515 PMCID: PMC7308495 DOI: 10.3389/fnana.2020.00037] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
Recent studies have suggested the neuroinvasive potential of severe acute respiratory coronavirus 2 (SARS-CoV-2). Notably, neuroinvasiveness might be involved in the pathophysiology of coronavirus disease 2019 (COVID-19). Some studies have demonstrated that synapse-connected routes may enable coronaviruses to access the central nervous system (CNS). However, evidence related to the presence of SARS-CoV-2 in the CNS, its direct impact on the CNS, and the contribution to symptoms suffered, remain sparse. Here, we review the current literature that indicates that SARS-CoV-2 can invade the nervous system. We also describe the neural circuits that are potentially affected by the virus and their possible role in the progress of COVID-19. In addition, we propose several strategies to understand, diagnose, and treat the neurological symptoms of COVID-19.
Collapse
Affiliation(s)
- Marcos F. DosSantos
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Medicina (Radiologia), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Brazil
| | - Sylvie Devalle
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Veronica Aran
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Daniela Capra
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Natália Roberta Roque
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Juliana de Mattos Coelho-Aguiar
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tânia Cristina Leite de Sampaio e Spohr
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Janice Gonçalves Subilhaga
- Setor de Pneumologia, Serviço de Clínica Médica, Hospital Federal dos Servidores do Estado, Rio de Janeiro, Brazil
| | - Cláudia Maria Pereira
- Programa de Pós-Graduação em Biomedicina Translacional e Odontologia Clínica e Experimental, Universidade do Grande Rio (Unigranrio), Duque de Caxias, Brazil
| | - Isabella D'Andrea Meira
- Departamento de Neurologia, Universidade Federal Fluminense, Niterói, Brazil
- Programa de Epilepsia do Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vivaldo Moura-Neto
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Neurociência Translacional, Instituto Nacional de Neurociência Translacional (INNT-UFRJ), Rio de Janeiro, Brazil
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Mungoven TJ, Meylakh N, Marciszewski KK, Macefield VG, Macey PM, Henderson LA. Microstructural changes in the trigeminal nerve of patients with episodic migraine assessed using magnetic resonance imaging. J Headache Pain 2020; 21:59. [PMID: 32471359 PMCID: PMC7260805 DOI: 10.1186/s10194-020-01126-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND There is histological evidence of microstructural changes in the zygomaticotemporal branch of the trigeminal nerve in migraineurs. This raises the possibility that altered trigeminal nerve properties contribute to migraine pathophysiology. Whilst it is not possible to explore the anatomy of small trigeminal nerve branches it is possible to explore the anatomy of the trigeminal root entry zone using magnetic resonance imaging in humans. The aim of this investigation is to assess the microstructure of the trigeminal nerve in vivo to determine if nerve alterations occur in individuals with episodic migraine. METHODS In 39 migraineurs and 39 matched controls, T1-weighted anatomical images were used to calculate the volume (mm3) and maximal cross-sectional area of the trigeminal nerve root entry zone; diffusion tensor images were used to calculate fractional anisotropy, mean diffusion, axial diffusion and radial diffusion. RESULTS There were significant differences between the left and right nerve of controls and migraineurs with respect to volume and not cross-sectional area. Migraineurs displayed reduced axial diffusion in the right nerve compared to the left nerve, and reduced fractional anisotropy in the left nerve compared to left controls. Furthermore, although there were no differences in mean diffusion or radial diffusion, regional analysis of the nerve revealed significantly greater radial diffusion in the middle and rostral portion of the left trigeminal nerve in migraineurs compared with controls. CONCLUSIONS Migraine pathophysiology is associated with microstructural abnormalities within the trigeminal nerve that are consistent with histological evidence of altered myelin and/or organization. These peripheral nerve changes may provide further insight into migraine pathophysiology and enable a greater understanding for targeted treatments of pain alleviation.
Collapse
Affiliation(s)
- Tiffani J Mungoven
- Department of Anatomy and Histology, F13, University of Sydney, Sydney, NSW, 2006, Australia
| | - Noemi Meylakh
- Department of Anatomy and Histology, F13, University of Sydney, Sydney, NSW, 2006, Australia
| | - Kasia K Marciszewski
- Department of Anatomy and Histology, F13, University of Sydney, Sydney, NSW, 2006, Australia
| | | | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, California, 90095, USA
| | - Luke A Henderson
- Department of Anatomy and Histology, F13, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
25
|
PKCγ interneurons, a gateway to pathological pain in the dorsal horn. J Neural Transm (Vienna) 2020; 127:527-540. [PMID: 32108249 DOI: 10.1007/s00702-020-02162-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/13/2020] [Indexed: 12/21/2022]
Abstract
Chronic pain is a frequent and disabling condition that is significantly maintained by central sensitization, which results in pathological amplification of responses to noxious and innocuous stimuli. As such, mechanical allodynia, or pain in response to a tactile stimulus that does not normally provoke pain, is a cardinal feature of chronic pain. Recent evidence suggests that the dorsal horn excitatory interneurons that express the γ isoform of protein kinase C (PKCγ) play a critical role in the mechanism of mechanical allodynia during chronic pain. Here, we review this evidence as well as the main aspects of the development, anatomy, electrophysiology, inputs, outputs, and pathophysiology of dorsal horn PKCγ neurons. Primary afferent high-threshold neurons transmit the nociceptive message to the dorsal horn of the spinal cord and trigeminal system where it activates second-order nociceptive neurons relaying the information to the brain. In physiological conditions, low-threshold mechanoreceptor inputs activate inhibitory interneurons in the dorsal horn, which may control activation of second-order nociceptive neurons. During chronic pain, low-threshold mechanoreceptor inputs now activate PKCγ neurons that forward the message to second-order nociceptive neurons, turning thus tactile inputs into pain. Several mechanisms may contribute to opening this gate, including disinhibition, activation of local astrocytes, release of diffusible factors such as reactive oxygen species, and alteration of the descending serotoninergic control on PKCγ neurons through 5-HT2A serotonin receptors. Dorsal horn PKCγ neurons, therefore, appear as a relevant therapeutic target to alleviate mechanical allodynia during chronic pain.
Collapse
|
26
|
Stinson C, Logan SM, Bellinger LL, Rao M, Kinchington PR, Kramer PR. Estradiol Acts in Lateral Thalamic Region to Attenuate Varicella Zoster Virus Associated Affective Pain. Neuroscience 2019; 414:99-111. [PMID: 31271831 DOI: 10.1016/j.neuroscience.2019.06.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
Abstract
Varicella zoster virus (VZV) results in chicken pox and herpes zoster. Female rats show a higher level of herpes zoster associated pain than males, consistent with human studies. In this study, we addressed the novel hypothesis that sex difference in herpes zoster associated pain is due, in part, to estradiol modulating activity in the thalamus. To test this hypothesis a high and low physiological dose of estradiol was administered to castrated and ovariectomized rats and the affective pain response was measured after injection of VZV into the whisker pad. Thalamic infusion of the estrogen receptor antagonist ICI 182,780 concomitant with a high dose of estradiol addressed the role of estradiol binding to its receptor to effect pain. Phosphorylated extracellular signal-regulated protein kinase (pERK) positive cells were measured in excitatory (glutaminase positive) and inhibitory (glutamate decarboxylase 67 positive) cells of the lateral thalamic region. Our results show that a high dose of estradiol significantly reduced the pain response in both males and females. pERK significantly increased in excitatory cells after treatment with a low dose of estradiol and increased in inhibitory cells after treatment with a high dose of estradiol. Administration of ICI 182,780 significantly increased the pain response, reduced expression of GABA related genes in the thalamic region and significantly reduced the number of inhibitory cells expressing pERK. The results suggest that estradiol attenuates herpes zoster pain by increasing the activity of inhibitory neurons within the thalamus and that this reduction includes an estrogen receptor dependent mechanism.
Collapse
Affiliation(s)
- Crystal Stinson
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States of America
| | - Shaun M Logan
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States of America
| | - Larry L Bellinger
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States of America
| | - Mahesh Rao
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States of America
| | - Paul R Kinchington
- Department of Ophthalmology and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Room 1020 EEI building 203 Lothrop Street, Pittsburgh, PA 15213, United States of America
| | - Phillip R Kramer
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States of America.
| |
Collapse
|
27
|
Kramer PR, Rao M, Stinson C, Bellinger LL, Kinchington PR, Yee MB. Aromatase Derived Estradiol Within the Thalamus Modulates Pain Induced by Varicella Zoster Virus. Front Integr Neurosci 2018; 12:46. [PMID: 30369871 PMCID: PMC6194186 DOI: 10.3389/fnint.2018.00046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/14/2018] [Indexed: 12/18/2022] Open
Abstract
Herpes zoster or shingles is the result of varicella zoster virus (VZV) infection and often results in chronic pain that lasts for months after visible symptoms subside. Testosterone often attenuates pain in males. Previous work demonstrates ovarian estrogen effects γ-aminobutyric acid (GABA) signaling in the thalamus, reducing pain but the role of testosterone within the thalamus is currently unknown. Because aromatase affects pain and is present in the thalamus we tested a hypothesis that testosterone converted to estrogen in the thalamus attenuates herpes zoster induced pain. To address this hypothesis, male Sprague-Dawley rats received whisker pad injection of either MeWo cells or MeWo cells containing VZV. To reduce aromatase derived estrogen in these animals we injected aromatase inhibitor letrozole systemically or infused it into the thalamus. To test if estrogen was working through the estrogen receptor (ER) agonist, 4, 4′, 4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) was infused concomitant with letrozole. Motivational and affective pain was measured after letrozole and/or PPT treatment. Vesicular GABA transporter (VGAT) is important in pain signaling. Because estrogen effects VGAT expression we measured its transcript and protein levels after letrozole treatment. Virus injection and letrozole significantly increased the pain response but thalamic infusion of PPT reduced zoster pain. Letrozole increased the number of thalamic neurons staining for phosphorylated ERK (pERK) but decreased VGAT expression. The results suggest in male rats aromatase derived estradiol interacts with the ER to increase VGAT expression and increase neuronal inhibition in the thalamus to attenuate VZV induced pain.
Collapse
Affiliation(s)
- Phillip R Kramer
- Department of Biomedical Science, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Mahesh Rao
- Department of Biomedical Science, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Crystal Stinson
- Department of Biomedical Science, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Larry L Bellinger
- Department of Biomedical Science, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Paul R Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, Eye and Ear Foundation, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael B Yee
- Department of Ophthalmology and of Molecular Microbiology and Genetics, Eye and Ear Foundation, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
28
|
Abstract
OBJECTIVES The aim of this study is to investigate the role of peroxisome proliferator-activated receptor-gamma isoform (PPARγ), in trigeminal neuropathic pain utilizing a novel mouse trigeminal inflammatory compression (TIC) injury model. RESULTS The study determined that the PPARγ nuclear receptor plays a significant role in trigeminal nociception transmission, evidenced by: 1) Intense PPARγ immunoreactivity is expressed 3 weeks after TIC nerve injury in the spinal trigeminal caudalis, the termination site of trigeminal nociceptive nerve fibers. 2) Systemic administration of a PPARγ agonist, pioglitazone (PIO), attenuates whisker pad mechanical allodynia at doses of 300 mg/kg i.p. and 600 mg/kg p.o. 3) Administration of a PPARγ antagonist, GW9662 (30 mg/kg i.p.), prior to providing the optimal dose of PIO (300 mg/kg i.p.) blocked the analgesic effect of PIO. DISCUSSION This is the first study localizing PPARγ immunoreactivity throughout the brainstem trigeminal sensory spinal nucleus (spV) and its increase three weeks after TIC nerve injury. This is also the first study to demonstrate that activation of PPARγ attenuates trigeminal hypersensitivity in the mouse TIC nerve injury model. The findings presented here suggest the possibility of utilizing the FDA approved diabetic treatment drug, PIO, as a new therapeutic that targets PPARγ for treatment of patients suffering from orofacial neuropathic pain.
Collapse
|
29
|
Van der Cruyssen F, Politis C. Neurophysiological aspects of the trigeminal sensory system: an update. Rev Neurosci 2018; 29:115-123. [DOI: 10.1515/revneuro-2017-0044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/20/2017] [Indexed: 02/04/2023]
Abstract
AbstractThe trigeminal system is one of the most complex cranial nerve systems of the human body. Research on it has vastly grown in recent years and concentrated more and more on molecular mechanisms and pathophysiology, but thorough reviews on this topic are lacking, certainly on the normal physiology of the trigeminal sensory system. Here we review the current literature on neurophysiology of the trigeminal nerve from peripheral receptors up to its central projections toward the somatosensory cortex. We focus on the most recent scientific discoveries and describe historical relevant research to substantiate further. One chapter on new insights of the pathophysiology of pain at the level of the trigeminal system is added. A database search of Medline, Embase and Cochrane was conducted with the search terms ‘animal study’, ‘neurophysiology’, ‘trigeminal’, ‘oral’ and ‘sensory’. Articles were manually selected after reading the abstract and where needed the article. Reference lists also served to include relevant research articles. Fifty-six articles were included after critical appraisal. Physiological aspects on mechanoreceptors, trigeminal afferents, trigeminal ganglion and central projections are reviewed in light of reference works. Embryologic and anatomic insights are cited where needed. A brief description of pathophysiology of pain pathways in the trigeminal area and recent advances in dental stem cell research are also discussed. Neurophysiology at the level of the central nervous system is not reviewed. The current body of knowledge is mainly based on animal and cadaveric studies, but recent advancements in functional imaging and molecular neuroscience are elucidating the pathways and functioning of this mixed nerve system. Extrapolation of animal studies or functioning of peripheral nerves should be warranted.
Collapse
|
30
|
Changes in the expression of voltage-gated sodium channels Nav1.3, Nav1.7, Nav1.8, and Nav1.9 in rat trigeminal ganglia following chronic constriction injury. Neuroreport 2018; 27:929-34. [PMID: 27327156 DOI: 10.1097/wnr.0000000000000632] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Voltage-gated sodium channels (VGSCs), especially the tetrodotoxin-sensitive Nav1.3 and Nav1.7, and the tetrodotoxin-resistant Nav1.8 and Nav1.9, have been implicated in acute and chronic neuropathic pain. The aim of this study was to investigate the expression of VGSC Nav1.3, Nav1.7, Nav1.8, and Nav1.9 after nerve injury and their roles in the development of trigeminal neuralgia (TN). We used the infraorbital nerve-chronic constriction injury model of TN in the rat. The time course of changes in the mechanical pain threshold was examined. In addition, real-time PCR and double immunofluorescence staining of VGSC α subunits were used to evaluate messenger RNA and protein expression, respectively, in the trigeminal ganglion. Behavioral tests showed that the mechanical pain threshold decreased significantly 4-42 days after surgery and reached the lowest observed value by day 12. Compared with sham-operated controls, we found that trigeminal ganglion in rats subjected to an infraorbital nerve-chronic constriction injury showed upregulation of Nav1.3 and downregulation of Nav1.7, Nav1.8, and Nav1.9 messenger RNA and protein levels. Our findings suggest that VGSC may participate in the regulation of TN.
Collapse
|
31
|
Xu G, Zhou C, Liu S, Li W, Tang W. Electrophysiological characteristics of the frontal nerve in patients with herpetic ophthalmic neuralgia. Muscle Nerve 2018; 57:973-980. [PMID: 29314073 DOI: 10.1002/mus.26057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2017] [Indexed: 11/06/2022]
Abstract
INTRODUCTION The aim of this study was to explore a method for obtaining sensory nerve action potentials (SNAPs) of the supratrochlear (STN) and supraorbital (SON) nerves and evaluate the function of affected nerves in patients with herpetic ophthalmic neuralgia (HON). METHODS Thirty healthy volunteers and 40 subjects with subacute HON participated in this study. RESULTS The amplitudes and sensory conduction velocities (SCVs) that predicted HON were identified. The corresponding cutoff values for the amplitudes ranged from 11.10 μV to 12.45 μV. The corresponding cutoff values for the SCVs ranged from 43.14 m/s to 44.64 m/s. SCVs were markedly lower on the affected side compared with healthy volunteers (P < 0.05), and the amplitudes of SNAPs on the affected side were decreased by 36% compared with healthy volunteers (P < 0.05). DISCUSSION SCVs of STN and SONs can be obtained with the 3-channel method and used to evaluate myelinated fibers in patients with HON. Muscle Nerve 57: 973-980, 2018.
Collapse
Affiliation(s)
- Gang Xu
- Department of Rehabilitation Medicine, Affiliated Tenth People's Hospital of Tongji University, Shanghai Tenth People's Hospital, 301 Middle Yanchang Road Shanghai, 200072, China
| | - Chaosheng Zhou
- Department of Rehabilitation Medicine, Affiliated Tenth People's Hospital of Tongji University, Shanghai Tenth People's Hospital, 301 Middle Yanchang Road Shanghai, 200072, China
| | - Shasha Liu
- Department of Rehabilitation Medicine, Affiliated Tenth People's Hospital of Tongji University, Shanghai Tenth People's Hospital, 301 Middle Yanchang Road Shanghai, 200072, China
| | - Wen Li
- Department of Rehabilitation Medicine, Affiliated Tenth People's Hospital of Tongji University, Shanghai Tenth People's Hospital, 301 Middle Yanchang Road Shanghai, 200072, China
| | - Weizhen Tang
- Department of Rehabilitation Medicine, Affiliated Tenth People's Hospital of Tongji University, Shanghai Tenth People's Hospital, 301 Middle Yanchang Road Shanghai, 200072, China
| |
Collapse
|
32
|
Feller L, Khammissa RAG, Fourie J, Bouckaert M, Lemmer J. Postherpetic Neuralgia and Trigeminal Neuralgia. PAIN RESEARCH AND TREATMENT 2017; 2017:1681765. [PMID: 29359044 PMCID: PMC5735631 DOI: 10.1155/2017/1681765] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/25/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023]
Abstract
Postherpetic neuralgia (PHN) is an unpredictable complication of varicella zoster virus- (VZV-) induced herpes zoster (HZ) which often occurs in elderly and immunocompromised persons and which can induce psychosocial dysfunction and can negatively impact on quality of life. Preventive options for PHN include vaccination of high-risk persons against HZ, early use of antiviral agents, and robust management of pain during the early stage of acute herpes zoster. If it does occur, PHN may persist for months or even years after resolution of the HZ mucocutaneous eruptions, and treatment is often only partially effective. Classical trigeminal neuralgia is a severe orofacial neuropathic pain condition characterized by unilateral, brief but recurrent, lancinating paroxysmal pain confined to the distribution of one or more of the branches of the trigeminal nerve. It may be idiopathic or causally associated with vascular compression of the trigeminal nerve root. The anticonvulsive agents, carbamazepine or oxcarbazepine, constitute the first-line treatment. Microvascular decompression or ablative procedures should be considered when pharmacotherapy is ineffective or intolerable. The aim of this short review is briefly to discuss the etiopathogenesis, clinical features, and treatment of PHN and classical trigeminal neuralgia.
Collapse
Affiliation(s)
- L. Feller
- Department of Periodontology and Oral Medicine, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - R. A. G. Khammissa
- Department of Periodontology and Oral Medicine, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - J. Fourie
- Department of Periodontology and Oral Medicine, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| | - M. Bouckaert
- Department of Maxillofacial and Oral Surgery, Sefako Makgatho University, Pretoria, South Africa
| | - J. Lemmer
- Department of Periodontology and Oral Medicine, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| |
Collapse
|
33
|
de Matos NM, Hock A, Wyss M, Ettlin DA, Brügger M. Neurochemical dynamics of acute orofacial pain in the human trigeminal brainstem nuclear complex. Neuroimage 2017; 162:162-172. [DOI: 10.1016/j.neuroimage.2017.08.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 01/25/2023] Open
|
34
|
Lopes DM, Denk F, McMahon SB. The Molecular Fingerprint of Dorsal Root and Trigeminal Ganglion Neurons. Front Mol Neurosci 2017; 10:304. [PMID: 29018326 PMCID: PMC5623188 DOI: 10.3389/fnmol.2017.00304] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/11/2017] [Indexed: 12/15/2022] Open
Abstract
The dorsal root ganglia (DRG) and trigeminal ganglia (TG) are clusters of cell bodies of highly specialized sensory neurons which are responsible for relaying information about our environment to the central nervous system. Despite previous efforts to characterize sensory neurons at the molecular level, it is still unknown whether those present in DRG and TG have distinct expression profiles and therefore a unique molecular fingerprint. To address this question, we isolated lumbar DRG and TG neurons using fluorescence-activated cell sorting from Advillin-GFP transgenic mice and performed RNA sequencing. Our transcriptome analyses showed that, despite being overwhelmingly similar, a number of genes are differentially expressed in DRG and TG neurons. Importantly, we identified 24 genes which were uniquely expressed in either ganglia, including an arginine vasopressin receptor and several homeobox genes, giving each population a distinct molecular fingerprint. We compared our findings with published studies to reveal that many genes previously reported to be present in neurons are in fact likely to originate from other cell types in the ganglia. Additionally, our neuron-specific results aligned well with a dataset examining whole human TG and DRG. We propose that the data can both improve our understanding of primary afferent biology and help contribute to the development of drug treatments and gene therapies which seek targets with unique or restricted expression patterns.
Collapse
Affiliation(s)
- Douglas M Lopes
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Franziska Denk
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Stephen B McMahon
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
35
|
Stinson C, Deng M, Yee MB, Bellinger LL, Kinchington PR, Kramer PR. Sex differences underlying orofacial varicella zoster associated pain in rats. BMC Neurol 2017; 17:95. [PMID: 28514943 PMCID: PMC5436469 DOI: 10.1186/s12883-017-0882-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 05/09/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Most people are initially infected with varicella zoster virus (VZV) at a young age and this infection results in chickenpox. VZV then becomes latent and reactivates later in life resulting in herpes zoster (HZ) or "shingles". Often VZV infects neurons of the trigeminal ganglia to cause ocular problems, orofacial disease and occasionally a chronic pain condition termed post-herpetic neuralgia (PHN). To date, no model has been developed to study orofacial pain related to varicella zoster. Importantly, the incidence of zoster associated pain and PHN is known to be higher in women, although reasons for this sex difference remain unclear. Prior to this work, no animal model was available to study these sex-differences. Our goal was to develop an orofacial animal model for zoster associated pain which could be utilized to study the mechanisms contributing to this sex difference. METHODS To develop this model VZV was injected into the whisker pad of rats resulting in IE62 protein expression in the trigeminal ganglia; IE62 is an immediate early gene in the VZV replication program. RESULTS Similar to PHN patients, rats showed retraction of neurites after VZV infection. Treatment of rats with gabapentin, an agent often used to combat PHN, ameliorated the pain response after whisker pad injection. Aversive behavior was significantly greater for up to 7 weeks in VZV injected rats over control inoculated rats. Sex differences were also seen such that ovariectomized and intact female rats given the lower dose of VZV showed a longer affective response than male rats. The phase of the estrous cycle also affected the aversive response suggesting a role for sex steroids in modulating VZV pain. CONCLUSIONS These results suggest that this rat model can be utilized to study the mechanisms of 1) orofacial zoster associated pain and 2) the sex differences underlying zoster associated pain.
Collapse
Affiliation(s)
- Crystal Stinson
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246 USA
| | - Mohong Deng
- Department of Oral and Maxillofacial Surgery, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People’s Republic of China
| | - Michael B Yee
- Dept Ophthalmology and of Molecular Microbiology and Genetics, 203 Lothrop St., Pittsburgh, PA 15213 USA
| | - Larry L. Bellinger
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246 USA
| | - Paul R. Kinchington
- Dept Ophthalmology and of Molecular Microbiology and Genetics, 203 Lothrop St., Pittsburgh, PA 15213 USA
| | - Phillip R. Kramer
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246 USA
| |
Collapse
|
36
|
DosSantos MF, Holanda-Afonso RC, Lima RL, DaSilva AF, Moura-Neto V. The role of the blood-brain barrier in the development and treatment of migraine and other pain disorders. Front Cell Neurosci 2014; 8:302. [PMID: 25339863 PMCID: PMC4189386 DOI: 10.3389/fncel.2014.00302] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/08/2014] [Indexed: 12/23/2022] Open
Abstract
The function of the blood-brain barrier (BBB) related to chronic pain has been explored for its classical role in regulating the transcellular and paracellular transport, thus controlling the flow of drugs that act at the central nervous system, such as opioid analgesics (e.g., morphine) and non-steroidal anti-inflammatory drugs. Nonetheless, recent studies have raised the possibility that changes in the BBB permeability might be associated with chronic pain. For instance, changes in the relative amounts of occludin isoforms, resulting in significant increases in the BBB permeability, have been demonstrated after inflammatory hyperalgesia. Furthermore, inflammatory pain produces structural changes in the P-glycoprotein, the major efflux transporter at the BBB. One possible explanation for these findings is the action of substances typically released at the site of peripheral injuries that could lead to changes in the brain endothelial permeability, including substance P, calcitonin gene-related peptide, and interleukin-1 beta. Interestingly, inflammatory pain also results in microglial activation, which potentiates the BBB damage. In fact, astrocytes and microglia play a critical role in maintaining the BBB integrity and the activation of those cells is considered a key mechanism underlying chronic pain. Despite the recent advances in the understanding of BBB function in pain development as well as its interference in the efficacy of analgesic drugs, there remain unknowns regarding the molecular mechanisms involved in this process. In this review, we explore the connection between the BBB as well as the blood-spinal cord barrier and blood-nerve barrier, and pain, focusing on cellular and molecular mechanisms of BBB permeabilization induced by inflammatory or neuropathic pain and migraine.
Collapse
Affiliation(s)
- Marcos F. DosSantos
- Universidade Federal do Rio de Janeiro – Campus MacaéRio de Janeiro, Brazil
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
- Headache and Orofacial Pain Effort, Department of Biologic and Materials Sciences and Michigan Center for Oral Health Research, School of Dentistry, University of MichiganAnn Arbor, MI, USA
| | - Rosenilde C. Holanda-Afonso
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Rodrigo L. Lima
- Departamento de Ortodontia e Odontopediatria, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de JaneiroBrazil
| | - Alexandre F. DaSilva
- Headache and Orofacial Pain Effort, Department of Biologic and Materials Sciences and Michigan Center for Oral Health Research, School of Dentistry, University of MichiganAnn Arbor, MI, USA
| | - Vivaldo Moura-Neto
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
- Instituto Estadual do Cérebro Paulo NiemeyerRio de Janeiro, Brazil
| |
Collapse
|
37
|
Peirs C, Patil S, Bouali-Benazzouz R, Artola A, Landry M, Dallel R. Protein kinase C gamma interneurons in the rat medullary dorsal horn: distribution and synaptic inputs to these neurons, and subcellular localization of the enzyme. J Comp Neurol 2014; 522:393-413. [PMID: 23818225 DOI: 10.1002/cne.23407] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 12/18/2022]
Abstract
The γ isoform of protein kinase C (PKCγ), which is concentrated in interneurons in the inner part of lamina II (IIi ) of the dorsal horn, has been implicated in the expression of tactile allodynia. Lamina IIi PKCγ interneurons were shown to be activated by tactile inputs and to participate in local circuits through which these inputs can reach lamina I, nociceptive output neurons. That such local circuits are gated by glycinergic inhibition and that A- and C-fibers low threshold mechanoreceptors (LTMRs) terminate in lamina IIi raise the general issue of synaptic inputs to lamina IIi PKCγ interneurons. Combining light and electron microscopic immunochemistry in the rat spinal trigeminal nucleus, we show that PKCγ-immunoreactivity is mostly restricted to interneurons in lamina IIi of the medullary dorsal horn, where they constitute 1/3 of total neurons. The majority of synapses on PKCγ-immunoreactive interneurons are asymmetric (likely excitatory). PKCγ-immunoreactive interneurons appear to receive exclusively myelinated primary afferents in type II synaptic glomeruli. Neither large dense core vesicle terminals nor type I synaptic glomeruli, assumed to be the endings of unmyelinated nociceptive terminals, were found on these interneurons. Moreover, there is no vesicular glutamate transporter 3-immunoreactive bouton, specific to C-LTMRs, on PKCγ-immunoreactive interneurons. PKCγ-immunoreactive interneurons contain GABAA ergic and glycinergic receptors. At the subcellular level, PKCγ-immunoreactivity is mostly concentrated on plasma membranes, close to, but not within, postsynaptic densities. That only myelinated primary afferents were found to contact PKCγ-immunoreactive interneurons suggests that myelinated, but not unmyelinated, LTMRs play a critical role in the expression of mechanical allodynia.
Collapse
Affiliation(s)
- Cédric Peirs
- Inserm/UdA U1107, Neuro-Dol: Trigeminal Pain and Migraine, Université d'Auvergne, Faculté de Chirurgie Dentaire, Clermont-Ferrand, 63000, France
| | | | | | | | | | | |
Collapse
|
38
|
Brain signature of chronic orofacial pain: a systematic review and meta-analysis on neuroimaging research of trigeminal neuropathic pain and temporomandibular joint disorders. PLoS One 2014; 9:e94300. [PMID: 24759798 PMCID: PMC3997345 DOI: 10.1371/journal.pone.0094300] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 03/15/2014] [Indexed: 12/02/2022] Open
Abstract
Brain neuroimaging has been widely used to investigate the bran signature of chronic orofacial pain, including trigeminal neuropathic pain (TNP) and pain related to temporomandibular joint disorders (TMD). We here systematically reviewed the neuroimaging literature regarding the functional and structural changes in the brain of TNP and TMD pain patients, using a computerized search of journal articles via PubMed. Ten TNP studies and 14 TMD studies were reviewed. Study quality and risk of bias were assessed based on the criteria of patient selection, the history of medication, the use of standardized pain/psychological assessments, and the model and statistics of imaging analyses. Qualitative meta-analysis was performed by examining the brain regions which showed significant changes in either brain functions (including the blood-oxygen-level dependent signal, cerebral blood flow and the magnetic resonance spectroscopy signal) or brain structure (including gray matter and white matter anatomy). We hypothesized that the neuroimaging findings would display a common pattern as well as distinct patterns of brain signature in the disorders. This major hypothesis was supported by the following findings: (1) TNP and TMD patients showed consistent functional/structural changes in the thalamus and the primary somatosensory cortex, indicating the thalamocortical pathway as the major site of plasticity. (2) The TNP patients showed more alterations at the thalamocortical pathway, and the two disorders showed distinct patterns of thalamic and insular connectivity. Additionally, functional and structural changes were frequently reported in the prefrontal cortex and the basal ganglia, suggesting the role of cognitive modulation and reward processing in chronic orofacial pain. The findings highlight the potential for brain neuroimaging as an investigating tool for understanding chronic orofacial pain.
Collapse
|
39
|
Li N, Lu ZY, Yu LH, Burnstock G, Deng XM, Ma B. Inhibition of G protein-coupled P2Y2 receptor induced analgesia in a rat model of trigeminal neuropathic pain. Mol Pain 2014; 10:21. [PMID: 24642246 PMCID: PMC3995183 DOI: 10.1186/1744-8069-10-21] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 03/04/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUNDS ATP and P2X receptors play important roles in the modulation of trigeminal neuropathic pain, while the role of G protein-coupled P2Y₂ receptors and the underlying mechanisms are less clear. The threshold and frequency of action potentials, fast inactivating transient K+ channels (IA) are important regulators of membrane excitability in sensory neurons because of its vital role in the control of the spike onset. In this study, pain behavior tests, QT-RT-PCR, immunohistochemical staining, and patch-clamp recording, were used to investigate the role of P2Y₂ receptors in pain behaviour. RESULTS In control rats: 1) UTP, an agonist of P2Y₂/P2Y₄ receptors, caused a significant decrease in the mean threshold intensities for evoking action potentials and a striking increase in the mean number of spikes evoked by TG neurons. 2) UTP significantly inhibited IA and the expression of Kv1.4, Kv3.4 and Kv4.2 subunits in TG neurons, which could be reversed by the P2 receptor antagonist suramin and the ERK antagonist U0126. In ION-CCI (chronic constriction injury of infraorbital nerve) rats: 1) mRNA levels of Kv1.4, Kv3.4 and Kv4.2 subunits were significantly decreased, while the protein level of phosphorylated ERK was significantly increased. 2) When blocking P2Y₂ receptors by suramin or injection of P2Y2R antisense oligodeoxynucleotides both led to a time- and dose-dependent reverse of allodynia in ION-CCI rats. 3) Injection of P2Y₂ receptor antisense oligodeoxynucleotides induced a pronounced decrease in phosphorylated ERK expression and a significant increase in Kv1.4, Kv3.4 and Kv4.2 subunit expression in trigeminal ganglia. CONCLUSIONS Our data suggest that inhibition of P2Y₂ receptors leads to down-regulation of ERK-mediated phosphorylation and increase of the expression of I(A)-related Kv channels in trigeminal ganglion neurons, which might contribute to the clinical treatment of trigeminal neuropathic pain.
Collapse
Affiliation(s)
| | | | | | | | - Xiao-ming Deng
- Department of Physiology, The Key Laboratory of Molecular Neurobiology, Ministry of Education, Second Military Medical University, Shanghai 200433, P,R, China.
| | | |
Collapse
|
40
|
Liang L, Diao Y, Xu Q, Zhang M. Transcranial segment of the trigeminal nerve: macro-/microscopic anatomical study using sheet plastination. Acta Neurochir (Wien) 2014; 156:605-12. [PMID: 24158246 DOI: 10.1007/s00701-013-1920-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Accepted: 10/09/2013] [Indexed: 01/07/2023]
Abstract
BACKGROUND Trigeminal neuralgia (TN) may be caused by the mechanical compression of the trigeminal nerve. In the studies on the location of mechanical irritation and entrapment of the nerve, attention has been paid mostly to vascular structures in the subarachnoid space. Few studies have explored the relationship between the trigeminal nerve and its surrounding structures along its course in the skull base. The aim of this study was to examine and trace the root, ganglion and three divisions of the trigeminal nerve and their relationships with surrounding soft and bony structures in the skull base, and to identify the likely mechanical compression points. METHODS A total of 26 adult cadavers (ten females, 16 males; age range, 45-81 years) were examined in this study, eight for dissection and 16 for sheet plastination study. RESULTS Anatomical structures that may make the trigeminal nerve susceptible to entrapment in the skull base were located at (1) the inferolateral edge of the mouth of Meckel's cave, (2) the middle cranial fossa dura and the lateral wall of the anterior intracavernous portion of the internal carotid artery, (3) the ridge of the medial wall of the foramen rotundum, and (4) the twisted periosteum and venous plexus of the foramen ovale. CONCLUSION This study identified four likely mechanical compression points along the course of the trigeminal nerve in the skull base. Knowledge of these TN-susceptible sites may be useful to both skull base surgeon and TN-animal model researcher, particularly when they study TN without vascular compression.
Collapse
Affiliation(s)
- Liang Liang
- Department of Anatomy, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
41
|
Wilcox SL, Gustin SM, Eykman EN, Fowler G, Peck CC, Murray GM, Henderson LA. Trigeminal Nerve Anatomy in Neuropathic and Non-neuropathic Orofacial Pain Patients. THE JOURNAL OF PAIN 2013; 14:865-72. [DOI: 10.1016/j.jpain.2013.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/13/2012] [Accepted: 02/28/2013] [Indexed: 01/21/2023]
|
42
|
Melin C, Jacquot F, Dallel R, Artola A. Segmental disinhibition suppresses C-fiber inputs to the rat superficial medullary dorsal horn via the activation of GABABreceptors. Eur J Neurosci 2012; 37:417-28. [DOI: 10.1111/ejn.12048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 09/13/2012] [Accepted: 09/27/2012] [Indexed: 12/17/2022]
Affiliation(s)
| | - Florian Jacquot
- Clermont Université; Université d'Auvergne; Neuro-Dol, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107; F-63001 Clermont-Ferrand; France
| | | | - Alain Artola
- Clermont Université; Université d'Auvergne; Neuro-Dol, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107; F-63001 Clermont-Ferrand; France
| |
Collapse
|
43
|
DosSantos MF, Martikainen IK, Nascimento TD, Love TM, Deboer MD, Maslowski EC, Monteiro AA, Vincent MB, Zubieta JK, DaSilva AF. Reduced basal ganglia μ-opioid receptor availability in trigeminal neuropathic pain: a pilot study. Mol Pain 2012; 8:74. [PMID: 23006894 PMCID: PMC3522528 DOI: 10.1186/1744-8069-8-74] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 09/07/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although neuroimaging techniques have provided insights into the function of brain regions involved in Trigeminal Neuropathic Pain (TNP) in humans, there is little understanding of the molecular mechanisms affected during the course of this disorder. Understanding these processes is crucial to determine the systems involved in the development and persistence of TNP. FINDINGS In this study, we examined the regional μ-opioid receptor (μOR) availability in vivo (non-displaceable binding potential BPND) of TNP patients with positron emission tomography (PET) using the μOR selective radioligand [11C]carfentanil. Four TNP patients and eight gender and age-matched healthy controls were examined with PET. Patients with TNP showed reduced μOR BPND in the left nucleus accumbens (NAc), an area known to be involved in pain modulation and reward/aversive behaviors. In addition, the μOR BPND in the NAc was negatively correlated with the McGill sensory and total pain ratings in the TNP patients. CONCLUSIONS Our findings give preliminary evidence that the clinical pain in TNP patients can be related to alterations in the endogenous μ-opioid system, rather than only to the peripheral pathology. The decreased availability of μORs found in TNP patients, and its inverse relationship to clinical pain levels, provide insights into the central mechanisms related to this condition. The results also expand our understanding about the impact of chronic pain on the limbic system.
Collapse
Affiliation(s)
- Marcos Fabio DosSantos
- Headache & Orofacial Pain Effort (H.O.P.E.), Department of Biologic and Materials Sciences and MCOHR, School of Dentistry, University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Federal do Rio de Janeiro, Brazil
| | - Ilkka Kristian Martikainen
- Headache & Orofacial Pain Effort (H.O.P.E.), Department of Biologic and Materials Sciences and MCOHR, School of Dentistry, University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
- Translational Neuroimaging Laboratory, Molecular and Behavioral Neuroscience Institute (MBNI), University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
| | - Thiago Dias Nascimento
- Headache & Orofacial Pain Effort (H.O.P.E.), Department of Biologic and Materials Sciences and MCOHR, School of Dentistry, University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
| | - Tiffany M Love
- Translational Neuroimaging Laboratory, Molecular and Behavioral Neuroscience Institute (MBNI), University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
| | - Misty Dawn Deboer
- Headache & Orofacial Pain Effort (H.O.P.E.), Department of Biologic and Materials Sciences and MCOHR, School of Dentistry, University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
| | - Eric C Maslowski
- UM3D Lab, Digital Media Commons/MLibrary, University of Michigan, Michigan, USA
| | - André Antonio Monteiro
- Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Federal do Rio de Janeiro, Brazil
| | - Maurice Borges Vincent
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Federal do Rio de Janeiro, Brazil
| | - Jon-Kar Zubieta
- Translational Neuroimaging Laboratory, Molecular and Behavioral Neuroscience Institute (MBNI), University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
| | - Alexandre F DaSilva
- Headache & Orofacial Pain Effort (H.O.P.E.), Department of Biologic and Materials Sciences and MCOHR, School of Dentistry, University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
- Translational Neuroimaging Laboratory, Molecular and Behavioral Neuroscience Institute (MBNI), University of Michigan, Michigan, Ann Arbor, MI 48109-5720, USA
| |
Collapse
|