1
|
Li X, Prudente AS, Prato V, Guo X, Hao H, Jones F, Figoli S, Mullen P, Wang Y, Tonello R, Lee SH, Shah S, Maffei B, Berta T, Du X, Gamper N. Peripheral gating of mechanosensation by glial diazepam binding inhibitor. J Clin Invest 2024; 134:e176227. [PMID: 38888973 PMCID: PMC11324294 DOI: 10.1172/jci176227] [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: 09/27/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
We report that diazepam binding inhibitor (DBI) is a glial messenger mediating crosstalk between satellite glial cells (SGCs) and sensory neurons in the dorsal root ganglion (DRG). DBI is highly expressed in SGCs of mice, rats, and humans, but not in sensory neurons or most other DRG-resident cells. Knockdown of DBI results in a robust mechanical hypersensitivity without major effects on other sensory modalities. In vivo overexpression of DBI in SGCs reduces sensitivity to mechanical stimulation and alleviates mechanical allodynia in neuropathic and inflammatory pain models. We further show that DBI acts as an unconventional agonist and positive allosteric modulator at the neuronal GABAA receptors, particularly strongly affecting those with a high-affinity benzodiazepine binding site. Such receptors are selectively expressed by a subpopulation of mechanosensitive DRG neurons, and these are also more enwrapped with DBI-expressing glia, as compared with other DRG neurons, suggesting a mechanism for a specific effect of DBI on mechanosensation. These findings identified a communication mechanism between peripheral neurons and SGCs. This communication modulates pain signaling and can be targeted therapeutically.
Collapse
Affiliation(s)
- Xinmeng Li
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Xianchuan Guo
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Han Hao
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Frederick Jones
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Sofia Figoli
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Pierce Mullen
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Yujin Wang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Shihab Shah
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Benito Maffei
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Xiaona Du
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
2
|
Antonopoulos SR, Durham PL. Grape seed extract suppresses calcitonin gene-related peptide secretion and upregulates expression of GAD 65/67 and GABAB receptor in primary trigeminal ganglion cultures. IBRO Neurosci Rep 2022; 13:187-197. [PMID: 36093283 PMCID: PMC9449751 DOI: 10.1016/j.ibneur.2022.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/20/2022] [Indexed: 12/02/2022] Open
Abstract
The trigeminal ganglion is implicated in the underlying pathology of migraine and temporomandibular joint disorders (TMD), which are orofacial pain conditions involving peripheral and central sensitization. The neuropeptide calcitonin gene-related peptide (CGRP) is synthesized in some trigeminal ganglion neurons, and its release promotes inflammation, peripheral and central sensitization, and pain signaling. Recent studies in preclinical migraine and TMD models provide evidence that dietary supplementation with grape seed extract (GSE) inhibits trigeminal pain signaling. The goal of this study was to investigate the cellular mechanisms by which GSE modulates primary trigeminal ganglion cultures. The effect of GSE on CGRP secretion was determined by radioimmunoassay. To determine if GSE effects involved modulation of CGRP or the GABAergic system, expression of CGRP, GAD 65 and 67, GABAA receptor, and GABAB1 and GABAB2 receptor subunits were investigated by immunocytochemistry. GSE significantly inhibited basal CGRP secretion but did not alter neuronal CGRP expression. GAD 65 and 67 expression levels in neurons were significantly increased in response to GSE. While GSE did not cause a change in the neuronal expression of GABAA, GSE significantly increased GABAB1 expression in neurons, satellite glial cells, and Schwann cells. GABAB2 expression was significantly elevated in satellite glia and Schwann cells. These findings support the notion that GSE inhibition of basal CGRP secretion involves increased neuronal GAD 65 and 67 and GABAB receptor expression. GSE repression of CGRP release coupled with increased GABAB1 and GABAB2 glial cell expression would be neuroprotective by suppressing neuronal and glial excitability in the trigeminal ganglion. Grape seed extract inhibited basal CGRP release from cultured trigeminal neurons Neuronal expression of GAD 65/67 and GABAB1 was stimulated by grape seed extract Grape seed extract also increased GABAB1 in satellite glial cells and Schwann cells Glial expression of G protein-coupled GABAB2 was enhanced by grape seed extract Grape seed extract promotes neuroprotective cellular changes in trigeminal ganglion
Collapse
|
3
|
Sivakumar D, Ramli R. GABAergic signalling in modulation of dental pain. Eur J Pharmacol 2022; 924:174958. [DOI: 10.1016/j.ejphar.2022.174958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
|
4
|
KC E, Islam J, Kim S, Kim HK, Park YS. Pain Relief in a Trigeminal Neuralgia Model via Optogenetic Inhibition on Trigeminal Ganglion Itself With Flexible Optic Fiber Cannula. Front Cell Neurosci 2022; 16:880369. [PMID: 35573830 PMCID: PMC9096083 DOI: 10.3389/fncel.2022.880369] [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: 02/21/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022] Open
Abstract
The trigeminal ganglion (TG) is the primary site of aberration in trigeminal neuralgia (TN), and hence a crucial site where afferent input can be modulated. Here, we postulated that inhibiting TG via optogenetics using flexible optic cannula would diminish brainstem trigeminal nucleus caudalis (TNC) neuronal activity and pain behavior in TN rat model. Infraorbital nerve constriction was employed to induce TN in female Sprague-Dawley rats, while naive and sham rats served as controls. TG-directed microinjections of AAV virus containing either the optogenetic or null vector were delivered to rats in each group. In vivo electrophysiological responses were obtained from the ventral posteromedial nucleus (VPm) of the thalamus with simultaneous TG optogenetic stimulation using flexible optic cannula as well the effects on behavioral responses were investigated. Recordings in TN rats revealed a decrease in burst firing activity during yellow laser driven inhibition on TG, as well as considerably improved behavioral responses. In contrast, we noticed persistent hypersensitivity and increased tonic firing with blue laser stimulation which indicates that TG inhibition can synchronize trigeminal pain signal transmission in a TN animal model. The potential of an optogenetic approach in TG itself with flexible optic fiber to directly disrupt the trigeminal pain circuitry delivers fundamental underpinnings toward its prospective as a trigeminal neuralgia management.
Collapse
Affiliation(s)
- Elina KC
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Jaisan Islam
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Soochong Kim
- Department of Veterinary Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea
| | - Hyong Kyu Kim
- Department of Medicine and Microbiology, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | - Young Seok Park
- Department of Medical Neuroscience, College of Medicine, Chungbuk National University, Cheongju, South Korea
- Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, South Korea
| |
Collapse
|
5
|
Korczeniewska OA, James MH, Eliav T, Katzmann Rider G, Mehr JB, Affendi H, Aston-Jones G, Benoliel R. Chemogenetic inhibition of trigeminal ganglion neurons attenuates behavioural and neural pain responses in a model of trigeminal neuropathic pain. Eur J Pain 2021; 26:634-647. [PMID: 34767278 DOI: 10.1002/ejp.1887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nerve injury can lead to ectopic activation of injured nociceptorsand central sensitization characterized by allodynia and hyperalgesia. Reduction in the activity of primary afferent neurons has been shown to be sufficient in alleviating peripherally generated pain. The cell bodies of such trigeminal nociceptors are located in the trigeminal ganglia (TG) with central processes that terminate in the brainstem trigeminal nucleus caudalis (TNC). The TG is therefore a strategic locus where afferent input can be manipulated. We hypothesized that chemogenetic inhibition of TG would suppress TNC neuronal activity and attenuate pain behaviour in a rat model of painful traumatic trigeminal neuropathy (PTTN). METHODS Trigeminal neuropathic pain was induced in adult male Sprague-Dawley rats (n = 24) via chronic constriction injury to the infraorbital nerve (ION-CCI). Naïve and sham rats were used as controls (n = 20/group). Rats within each group received TG-directed microinjections of AAV virus containing either the inhibitory hM4Di-DREADD construct or EGFP. RESULTS In the ION-CCI group, systemic administration of the DREADD agonist clozapine N-oxide (CNO) reversed the hypersensitivity phenotype in animals expressing hM4Di but not EGFP. CNO-mediated activation of hM4Di DREADD in ION-CCI animals was also associated with reduced Fos expression in the TNC elicited by repeated mechanical stimulation of the dermatome ipsilateral to the injury. There was no effect of CNO on pain behaviour or TNC Fos expression in eGFP animals. CONCLUSION Our results indicate that DREADDs may offer an effective therapeutic approach for treatment of trigeminal neuropathic pain. SIGNIFICANCE Trigeminal neuropathic pain is highly resistant to therapy and we are in dire need of novel approaches. This study provides further evidence for the successful application of DREADDs as an effective tool for modulating central nervous system function. CNO mediated activation of hM4Di-DREADDs in the trigeminal ganglion (TG) attenuates nerve injury induced neuropathic pain by acting on hyperactive TG cells. It also establishes the TG as an effective target to manage pain in the face and head. Accessing the TG in clinical populations is a relatively simple and safe procedure, making this approach highly significant. Moreover, the methodology described here has applications in trigeminal neuropathic pain from traumatic other etiologies and in spinal neuropathic pain. Chronic pain syndromes are characterized by a progressive failure of brain centers to adequately inhibit pain and as these are identified, we may be able to target them for therapy. Therefore, our findings might have wide application in chronic pain syndromes.
Collapse
Affiliation(s)
- Olga A Korczeniewska
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers University, Newark, New Jersey, USA.,Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.,Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Tali Eliav
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Giannina Katzmann Rider
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers University, Newark, New Jersey, USA
| | - Jacqueline B Mehr
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.,Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Hafsa Affendi
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers University, Newark, New Jersey, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.,Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Rafael Benoliel
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers University, Newark, New Jersey, USA.,Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
6
|
Bhansali D, Teng SL, Lee CS, Schmidt BL, Bunnett NW, Leong KW. Nanotechnology for Pain Management: Current and Future Therapeutic Interventions. NANO TODAY 2021; 39:101223. [PMID: 34899962 PMCID: PMC8654201 DOI: 10.1016/j.nantod.2021.101223] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pain is one of the most common medical conditions and affects more Americans than diabetes, heart disease, and cancer combined. Current pain treatments mainly rely on opioid analgesics and remain unsatisfactory. The life-threatening side effects and addictive properties of opioids demand new therapeutic approaches. Nanomedicine may be able to address these challenges as it allows for sensitive and targeted treatments without some of the burdens associated with current clinical pain therapies. This review discusses the physiology of pain, the current landscape of pain treatment, novel targets for pain treatment, and recent and ongoing efforts to effectively treat pain using nanotechnology-based approaches. We highl ight advances in nanoparticle-based drug delivery to reduce side effects, gene therapy to tackle the source of pain, and nanomaterials-based scavenging to proactively mediate pain signaling.
Collapse
Affiliation(s)
- Divya Bhansali
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Shavonne L. Teng
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone School of Medicine, New York, NY 10010
| | - Caleb S. Lee
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Brian L. Schmidt
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, NY 10010
| | - Nigel W. Bunnett
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone School of Medicine, New York, NY 10010
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
- Department of Systems Biology, Columbia University, New York, NY 10027
| |
Collapse
|
7
|
Attenuation of Sensory Transmission Through the Rat Trigeminal Ganglion by GABA Receptor Activation. Neuroscience 2021; 471:80-92. [PMID: 34311018 DOI: 10.1016/j.neuroscience.2021.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/02/2021] [Accepted: 07/17/2021] [Indexed: 11/23/2022]
Abstract
While the trigeminal ganglion is often considered a passive conduit of sensory transmission, neurons and satellite glial cells (SGCs) within it can release neurotransmitters and express neuroreceptors. Some trigeminal ganglion neurons contain the neurotransmitter γ-aminobutyric acid (GABA) and express GABA receptors. There is behavioral evidence that increased GABA levels in the trigeminal ganglion decreases nociception, while a loss of GABA receptors results in hyperalgesia, although the neural mechanisms for this remain to be investigated. In this study, the expression of GABA receptors by trigeminal ganglion neurons that innervate rat labial skin and masseter muscle was compared using immunohistochemistry. The effect of intraganglionic administration of GABA receptor agonists was investigated by single unit recording of trigeminal brainstem and ganglion neuron responses to stimulation of the labial skin and/or masseter muscle in anesthetized rats. The mean frequency of expression of GABAA and GABAB receptors by masseter and labial skin ganglion neurons was 62.5% and 92.7%, and 55.4% and 20.3%, respectively. The expression of both GABA receptors was significantly greater in skin ganglion neurons. Masticatory muscle evoked brainstem trigeminal neuron responses were significantly attenuated by intraganglionic injection of muscimol (GABAA) but not baclofen (GABAB). The mechanical sensitivity of slow and fast conducting masticatory muscle afferent fibers was decreased and increased, respectively, by intraganglionic injection of both muscimol and baclofen. Activation of GABAA receptors may exert a gating effect on sensory transmission through the trigeminal ganglion by decreasing putative nociceptive input and enhancing innocuous sensory input.
Collapse
|
8
|
Tzeng HR, Lee MT, Fan PC, Knutson DE, Lai TH, Sieghart W, Cook J, Chiou LC. α6GABA A Receptor Positive Modulators Alleviate Migraine-like Grimaces in Mice via Compensating GABAergic Deficits in Trigeminal Ganglia. Neurotherapeutics 2021; 18:569-585. [PMID: 33111258 PMCID: PMC8116449 DOI: 10.1007/s13311-020-00951-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2020] [Indexed: 11/29/2022] Open
Abstract
Migraine is caused by hyperactivity of the trigeminovascular system, where trigeminal ganglia (TG) play an important role. This hyperactivity might originate from an underfunctional GABAergic system in TG. To investigate this possibility, we adapted a mouse model of migraine by inducing migraine-like grimaces in male mice via repeated injections of nitroglycerin (NTG, 10 mg/kg, i.p.), once every 2 days, for up to 5 sessions. Migraine-like facial pain scores were measured using the mouse grimace scale. Repeated NTG treatments in mice caused significant increases in migraine-like grimaces that were aborted and prevented by two anti-migraine agents sumatriptan and topiramate, respectively. After 5 sessions of NTG injections, the GABA-synthesizing enzyme, 65-kDa glutamate decarboxylase (GAD65), but not the GABA transporter 1 (GAT1) or the α6 subunit-containing GABAA receptors (α6GABAARs), was downregulated in mouse TG tissues. Taking advantage of the unaffected TG α6GABAAR expression in NTG-treated mice, we demonstrated that an α6GABAAR-selective positive allosteric modulator (PAM), DK-I-56-1, exhibited both abortive and prophylactic effects, comparable to those of sumatriptan and topiramate, respectively, in this migraine-mimicking mouse model. The brain-impermeable furosemide significantly prevented the effects of DK-I-56-1, suggesting its peripheral site of action, likely via preventing α6GABAAR modulation in TG. Results suggest that a decreased GABA synthesis caused by the reduced GAD65 expression in TG contributes to the trigeminovascular activation in this repeated NTG-induced migraine-mimicking model and that the unaltered α6GABAARs in TG are potential targets for migraine treatment. Thus, α6GABAAR-selective PAMs are potential anti-migraine agents for both abortive and preventive therapies.
Collapse
Affiliation(s)
- Hung-Ruei Tzeng
- Department of Pharmacology, Graduate Institute of Pharmacology College of Medicine, National Taiwan University, No. 1, Jen-Ai Rd., Section 1, Taipei, 10051, Taiwan
| | - Ming Tatt Lee
- Graduate Institute of Brain and Mind Sciences College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Faculty of Pharmaceutical Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Pi-Chuan Fan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, 10002, Taiwan
| | - Daniel E Knutson
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Tzu-Hsuan Lai
- Department of Pediatrics, National Taiwan University Hospital, Taipei, 10002, Taiwan
| | - Werner Sieghart
- Center for Brain Research, Department of Molecular Neurosciences, Medical University Vienna, 1090, Vienna, Austria
| | - James Cook
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Lih-Chu Chiou
- Department of Pharmacology, Graduate Institute of Pharmacology College of Medicine, National Taiwan University, No. 1, Jen-Ai Rd., Section 1, Taipei, 10051, Taiwan.
- Graduate Institute of Brain and Mind Sciences College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
9
|
Kim D, Kim KR, Kwon Y, Kim M, Kim MJ, Sim Y, Ji H, Park JJ, Cho JH, Choi H, Kim S. AAV-Mediated Combination Gene Therapy for Neuropathic Pain: GAD65, GDNF, and IL-10. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:473-483. [PMID: 32728596 PMCID: PMC7378317 DOI: 10.1016/j.omtm.2020.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022]
Abstract
Neuropathic pain is a chronic pain state characterized by nerve damage, inflammation, and nociceptive neuron hyperactivity. As the underlying pathophysiology is complex, a more effective therapy for neuropathic pain would be one that targets multiple elements. Here, we generated recombinant adeno-associated viruses (AAVs) encoding three therapeutic genes, namely, glutamate decarboxylase 65, glial cell-derived neurotrophic factor, and interleukin-10, with various combinations. The efficacy for pain relief was evaluated in a rat spared nerve injury model of neuropathic pain. The maximal analgesic effect was achieved when the AAVs expressing all three genes were administered to rats with neuropathic pain. The combination of two virus constructs expressing the three genes was named KLS-2031 and evaluated as a potential novel therapeutic for neuropathic pain. Single transforaminal epidural injections of KLS-2031 into the intervertebral foramen to target the appropriate dorsal root ganglion produced notable long-term analgesic effects in female and male rats. Furthermore, KLS-2031 mitigated the neuroinflammation, neuronal cell death, and dorsal root ganglion hyperexcitability induced by the spared nerve injury. These results suggest that KLS-2031 represents a promising therapeutic option for refractory neuropathic pain.
Collapse
Affiliation(s)
- Daewook Kim
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Kyung-Ran Kim
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Yejin Kwon
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Minjung Kim
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Min-Ju Kim
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Yeomoon Sim
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Hyelin Ji
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Jang-Joon Park
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Jong-Ho Cho
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Heonsik Choi
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Sujeong Kim
- Institute of BioInnovation Research, Kolon Life Science, 110 Magokdong-ro, Gangseo-gu, Seoul 07793, Republic of Korea
| |
Collapse
|
10
|
Kanao-Kanda M, Kanda H, Liu S, Roy S, Toborek M, Hao S. Viral Vector-Mediated Gene Transfer of Glutamic Acid Decarboxylase for Chronic Pain Treatment: A Literature Review. Hum Gene Ther 2020; 31:405-414. [PMID: 32041431 DOI: 10.1089/hum.2019.359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chronic pain is long-lasting nociceptive state, impairing the patient's quality of life. Existing analgesics are generally not effective in the treatment of chronic pain, some of which such as opioids have the risk of tolerance/dependence and overdose death with higher daily opioid doses for increasing analgesic effect. Opioid use disorders have already reached an epidemic level in the United States; therefore, nonopioid analgesic approach and/or use of nonpharmacologic interventions will be employed with increasing frequency. Viral vector-mediated gene therapy is promising in clinical trials in the nervous system diseases. Glutamic acid decarboxylase (GAD) enzyme, a key enzyme in biosynthesis of γ-aminobutyric acid (GABA), plays an important role in analgesic mechanism. In the literature review, we used PubMed and bioRxiv to search the studies, and the eligible criteria include (1) article written in English, (2) use of viral vectors expressing GAD67 or GAD65, and (3) preclinical pain models. We identified 13 eligible original research articles, in which the pain models include nerve injury, HIV-related pain, painful diabetic neuropathy, and formalin test. GAD expressed by the viral vectors from all the reports produced antinociceptive effects. Restoring GABA systems is a promising therapeutic strategy for chronic pain, which provides evidence for the clinical trial of gene therapy for pain in the near future.
Collapse
Affiliation(s)
- Megumi Kanao-Kanda
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Hirotsugu Kanda
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida.,Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Sabita Roy
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Michal Toborek
- Department of Anesthesiology & Critical Care Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
11
|
Bista P, Imlach WL. Pathological Mechanisms and Therapeutic Targets for Trigeminal Neuropathic Pain. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E91. [PMID: 31443547 PMCID: PMC6789505 DOI: 10.3390/medicines6030091] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/31/2022]
Abstract
Trigeminal neuropathic pain is a chronic pain condition caused by damage or inflammation of the trigeminal nerve or its branches, with both peripheral and central nervous system dysfunction contributing to the disorder. Trigeminal pain conditions present with diagnostic and therapeutic challenges to healthcare providers and often require multiple therapeutic approaches for pain reduction. This review will provide the overview of pathophysiology in peripheral and central nociceptive circuits that are involved in neuropathic pain conditions involving the trigeminal nerve and the current therapeutics that are used to treat these disorders. Recent advances in treatment of trigeminal pain, including novel therapeutics that target ion channels and receptors, gene therapy and monoclonal antibodies that have shown great promise in preclinical studies and clinical trials will also be described.
Collapse
Affiliation(s)
- Pawan Bista
- Department of Physiology & Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Wendy L Imlach
- Department of Physiology & Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
12
|
Fan W, Zhu X, He Y, Zhu M, Wu Z, Huang F, He H. The role of satellite glial cells in orofacial pain. J Neurosci Res 2018; 97:393-401. [PMID: 30450738 DOI: 10.1002/jnr.24341] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/29/2018] [Accepted: 09/21/2018] [Indexed: 12/14/2022]
Abstract
Some chronic pain conditions in the orofacial region are common, the mechanisms underlying which are unresolved. Satellite glial cells (SGCs) are the glial cells of the peripheral nervous system. In the sensory ganglia, each neuronal body is surrounded by SGCs forming distinct functional units. The unique structural organization enables SGCs to communicate with each other and with their enwrapped neurons via a variety of ways. There is a growing body of evidence that SGCs can influence the level of neuronal excitability and are involved in the development and/or maintenance of pain. The aim of this review was to summarize the latest advances made about the implication of SGCs in orofacial pain. It may offer new targets for the development of orofacial pain treatment.
Collapse
Affiliation(s)
- Wenguo Fan
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China.,Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiao Zhu
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Yifan He
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Mengzhu Zhu
- Department of Rheumatology, Chinese Medicine Hospital in Linyi City, Shandong, China
| | - Zhi Wu
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Qiao LN, Liu JL, Tan LH, Yang HL, Zhai X, Yang YS. Effect of electroacupuncture on thermal pain threshold and expression of calcitonin-gene related peptide, substance P and γ-aminobutyric acid in the cervical dorsal root ganglion of rats with incisional neck pain. Acupunct Med 2017; 35:276-283. [PMID: 28600329 PMCID: PMC5561363 DOI: 10.1136/acupmed-2016-011177] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2017] [Indexed: 02/06/2023]
Abstract
Objective Acupuncture therapy effectively reduces post-surgical pain, but its mechanism of action remains unclear. The aim of this study was to investigate whether expression of γ-aminobutyric acid (GABA) and the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) in the primary sensory neurons of cervical dorsal root ganglia (DRG) are involved in electroacupuncture (EA)-induced analgesia in a rat model of incisional neck pain. Methods The pain model was established by making a longitudinal midline neck incision in 60 rats. Another 15 rats underwent sham surgery (normal group). Post-incision, 15 rats remained untreated (model group) and 45 rats underwent EA (frequency 2/100 Hz, intensity 1 mA) at bilateral LI18, LI4-PC6 or ST36-GB34 (n=15 each) for 30 min at 4 hours, 24 hours, and 48 hours post-surgery, followed by thermal pain threshold (PT) measurement. 30 min later, the rats were euthanased and cervical (C3-6) DRGs removed for measurement of immunoreactivity and mRNA expression of SP/CGRP and the GABAergic neuronal marker glutamic acid decarboxylase 67 (GAD67). Results Thermal PT was significantly lower in the model group versus the normal group and increased in the LI18 and LI4-PC6 groups but not the ST36-GB34 group compared with the model group. Additionally, EA at LI18 and LI4-PC6 markedly suppressed neck incision-induced upregulation of mRNA/protein expression of SP/CGRP, and upregulated mRNA/protein expression of GAD67 in the DRGs of C3-6 segments. Conclusions EA at LI18/LI4-PC6 increases PT in rats with incisional neck pain, which is likely related to downregulation of pronociceptive mediators SP/CGRP and upregulation of the inhibitory transmitter GABA in the primary sensory neurons of cervical DRGs.
Collapse
Affiliation(s)
- Li-Na Qiao
- Department of Biochemistry and Molecular Biology, Institute of Acu-Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun-Ling Liu
- Department of Physiology, Institute of Acu-Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lian-Hong Tan
- Department of Biochemistry and Molecular Biology, Institute of Acu-Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hai-Long Yang
- Department of Biochemistry and Molecular Biology, Institute of Acu-Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China.,Institute of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xu Zhai
- Department of Biochemistry and Molecular Biology, Institute of Acu-Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong-Sheng Yang
- Department of Biochemistry and Molecular Biology, Institute of Acu-Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Xie AX, Lee JJ, McCarthy KD. Ganglionic GFAP + glial Gq-GPCR signaling enhances heart functions in vivo. JCI Insight 2017; 2:e90565. [PMID: 28138563 DOI: 10.1172/jci.insight.90565] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The sympathetic nervous system (SNS) accelerates heart rate, increases cardiac contractility, and constricts resistance vessels. The activity of SNS efferent nerves is generated by a complex neural network containing neurons and glia. Gq G protein-coupled receptor (Gq-GPCR) signaling in glial fibrillary acidic protein-expressing (GFAP+) glia in the central nervous system supports neuronal function and regulates neuronal activity. It is unclear how Gq-GPCR signaling in GFAP+ glia affects the activity of sympathetic neurons or contributes to SNS-regulated cardiovascular functions. In this study, we investigated whether Gq-GPCR activation in GFAP+ glia modulates the regulatory effect of the SNS on the heart; transgenic mice expressing Gq-coupled DREADD (designer receptors exclusively activated by designer drugs) (hM3Dq) selectively in GFAP+ glia were used to address this question in vivo. We found that acute Gq-GPCR activation in peripheral GFAP+ glia significantly accelerated heart rate and increased left ventricle contraction. Pharmacological experiments suggest that the glial-induced cardiac changes were due to Gq-GPCR activation in satellite glial cells within the sympathetic ganglion; this activation led to increased norepinephrine (NE) release and beta-1 adrenergic receptor activation within the heart. Chronic glial Gq-GPCR activation led to hypotension in female Gfap-hM3Dq mice. This study provides direct evidence that Gq-GPCR activation in peripheral GFAP+ glia regulates cardiovascular functions in vivo.
Collapse
|
15
|
Long H, Liao L, Zhou Y, Shan D, Gao M, Huang R, Yang X, Lai W. A novel technique of delivering viral vectors to trigeminal ganglia in rats. Eur J Oral Sci 2017; 125:1-7. [PMID: 28067416 DOI: 10.1111/eos.12326] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Hu Long
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Lina Liao
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Yang Zhou
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Di Shan
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Meiya Gao
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Renhuan Huang
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Xin Yang
- Department of Stomatology; Shanghai Tenth People's Hospital; Tongji University School of Medicine; Shanghai China
| | - Wenli Lai
- Department of Orthodontics; State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| |
Collapse
|
16
|
Hu C, Cai Z, Lu Y, Cheng X, Guo Q, Wu Z, Zhang Q. Nonviral vector plasmid DNA encoding human proenkephalin gene attenuates inflammatory and neuropathic pain-related behaviors in mice. Neurosci Lett 2016; 634:87-93. [PMID: 27693568 DOI: 10.1016/j.neulet.2016.09.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/17/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022]
Abstract
Inflammatory pain and neuropathic pain are major clinical health issues that represent considerable social and economic burden worldwide. In the present study, we investigated the anti-nociceptive efficacy of delivery of human proenkephalin gene by a plasmid DNA vector (pVAX1-PENK) on complete Freund's adjuvant (CFA) induced inflammatory pain and spared nerve injury (SNI) induced neuropathic pain in mice. Mice were intramuscularly or intrathecally administered pVAX1 or pVAX1-PENK, respectively. Pain thresholds in the pVAX1-PENK treated mice were significantly higher at day 3, then reached a peak at day 7 and lasted until day 28 after gene transfer, and the analgesic effect of pVAX1-PENK was blocked with naloxone hydrochloride. In contrast, pVAX1 treated mice did not significantly improve pain thresholds. These results indicate that peripheral or spinal delivery of a plasmid encoding human proenkephalin gene provides a potential therapeutic strategy for inflammatory pain and neuropathic pain.
Collapse
Affiliation(s)
- Chunsheng Hu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China; College of Life Science and Bioengineering, Beijing University of Technology, Beijing 10024, People's Republic of China
| | - Zhenzhen Cai
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Yuxin Lu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Xiaochen Cheng
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Qi Guo
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | - Zuze Wu
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China; College of Life Science and Bioengineering, Beijing University of Technology, Beijing 10024, People's Republic of China
| | - Qinglin Zhang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China.
| |
Collapse
|
17
|
Guo C, Li M, Qi X, Lin G, Cui F, Li F, Wu X. Intranasal delivery of nanomicelle curcumin promotes corneal epithelial wound healing in streptozotocin-induced diabetic mice. Sci Rep 2016; 6:29753. [PMID: 27405815 PMCID: PMC5378915 DOI: 10.1038/srep29753] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/23/2016] [Indexed: 12/22/2022] Open
Abstract
Corneal nerves are mainly derived from the ophthalmic branch of the trigeminal ganglion (TG). Corneal neuropathy contributes to epithelial degenerative changes in diabetic keratopathy. Efficient drug delivery to TG may be beneficial for the treatment of diabetic keratopathy. This article described intranasal delivery of nanomicelle curcumin to correct pathophysiological conditions in TG to promote corneal epithelial/nerve wound healing in streptozotocin-induced diabetic mice. A diabetic mice model with corneal epithelium abrasion was established. Ocular topical and/or intranasal nanomicelle curcumin treatments were performed, and treatment efficacy and mechanisms of action were explored. Results showed that intranasal nanomicelle curcumin treatment promoted corneal epithelial wound healing and recovery of corneal sensation. Enhanced accumulation of reactive oxygen species, reduced free radical scavengers, increased mRNA expressions of inflammatory cytokines, and decreased mRNA expressions of neurotrophic factors in the cornea and TG neuron were observed in diabetic mice with corneal epithelium abrasions. Intranasal nanomicelle curcumin treatment effectively recovered these pathophysiological conditions, especially that of the TG neuron, and a strengthened recovery was observed with ocular topical combined with intranasal treatment. These findings indicated that intranasal curcumin treatment effectively helped promote diabetic corneal epithelial/nerve wound healing. This novel treatment might be a promising strengthened therapy for diabetic keratopathy.
Collapse
Affiliation(s)
- Chuanlong Guo
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Mengshuang Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan, Jinan 250022, China
| | - Xia Qi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Guiming Lin
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan, Jinan 250022, China
| | - Fenghua Cui
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Fengjie Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
| | - Xianggen Wu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao 266071, China
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan, Jinan 250022, China
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
18
|
Crowley T, Cryan JF, Downer EJ, O'Leary OF. Inhibiting neuroinflammation: The role and therapeutic potential of GABA in neuro-immune interactions. Brain Behav Immun 2016; 54:260-277. [PMID: 26851553 DOI: 10.1016/j.bbi.2016.02.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/22/2016] [Accepted: 02/02/2016] [Indexed: 12/25/2022] Open
Abstract
The central nervous system, once thought to be a site of immunological privilege, has since been found to harbour immunocompetent cells and to communicate with the peripheral nervous system. In the central nervous system (CNS), glial cells display immunological responses to pathological and physiological stimuli through pro- and anti-inflammatory cytokine and chemokine signalling, antigen presentation and the clearing of cellular debris through phagocytosis. While this neuroinflammatory signalling can act to reduce neuronal damage and comprises a key facet of CNS homeostasis, persistent inflammation or auto-antigen-mediated immunoreactivity can induce a positive feedback cycle of neuroinflammation that ultimately results in necrosis of glia and neurons. Persistent neuroinflammation has been recognised as a major pathological component of virtually all neurodegenerative diseases and has also been a focus of research into the pathology underlying psychiatric disorders. Thus, pharmacological strategies to curb the pathological effects of persistent neuroinflammation are of interest for many disorders of the CNS. Accumulating evidence suggests that GABAergic activities are closely bound to immune processes and signals, and thus the GABAergic neurotransmitter system might represent an important therapeutic target in modulating neuroinflammation. Here, we review evidence that inflammation induces changes in the GABA neurotransmitter system in the CNS and that GABAergic signalling exerts a reciprocal influence over neuroinflammatory processes. Together, the data support the hypothesis that the GABA system is a potential therapeutic target in the modulation of central inflammation.
Collapse
Affiliation(s)
- Tadhg Crowley
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Eric J Downer
- School of Medicine, Discipline of Physiology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
19
|
Ma F, Wang C, Yoder WE, Westlund KN, Carlson CR, Miller CS, Danaher RJ. Efficacy of Herpes Simplex Virus Vector Encoding the Human Preproenkephalin Gene for Treatment of Facial Pain in Mice. J Oral Facial Pain Headache 2016; 30:42-50. [PMID: 26817032 DOI: 10.11607/ofph.1512] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS To determine whether herpes simplex virus-based vectors can efficiently transduce mouse trigeminal ganglion (TG) neurons and attenuate preexisting nerve injury-induced whisker pad mechanical hypersensitivity in a trigeminal inflammatory compression (TIC) neuropathic pain model. METHODS Tissue transduction efficiencies of replication-conditional and replication-defective vectors to mouse whisker pads after topical administration and subcutaneous injection were assessed using quantitative real-time PCR (qPCR). Tissue tropism and transgene expression were assessed using qPCR and reverse-transcriptase qPCR following topical application of the vectors. Whisker pad mechanical sensitivities of TIC-injured mice were determined using graduated von Frey fibers before and after application of human preproenkephalin expressing replication-conditional vector (KHPE). Data were analyzed using one-way analysis of variance (ANOVA) and post hoc tests. RESULTS Transduction of target TGs was 8- to 50-fold greater after topical application than subcutaneous injection and ≥ 100-fold greater for replication-conditional than replication-defective vectors. Mean KHPE loads remained constant in TGs (4.5-9.8 × 10(4) copies/TG) over 3 weeks but were below quantifiable levels (10 copies/tissue) within 2 weeks of application in other nontarget cephalic tissues examined. Transgene expression in TGs was maximal during 2 weeks after topical application (100-200 cDNA copies/mL) and was below quantifiable levels (1 cDNA copy/mL) in all nontarget tissues. Topical KHPE administration reduced TIC-related mechanical hypersensitivity on whisker pads 4-fold (P < .05) for at least 1 week. CONCLUSION Topically administered KHPE produced a significant antinociceptive effect in the TIC mouse model of chronic facial neuropathic pain. This is the first report in which a gene therapeutic approach reduced trigeminal pain-related behaviors in an established pain state in mice.
Collapse
|
20
|
Kami K, Taguchi Ms S, Tajima F, Senba E. Improvements in impaired GABA and GAD65/67 production in the spinal dorsal horn contribute to exercise-induced hypoalgesia in a mouse model of neuropathic pain. Mol Pain 2016; 12:12/0/1744806916629059. [PMID: 27030712 PMCID: PMC4956002 DOI: 10.1177/1744806916629059] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/04/2015] [Indexed: 11/16/2022] Open
Abstract
Background Physical exercise effectively attenuates neuropathic pain, and multiple events including the inhibition of activated glial cells in the spinal dorsal horn, activation of the descending pain inhibitory system, and reductions in pro-inflammatory cytokines in injured peripheral nerves may contribute to exercise-induced hypoalgesia. Since fewer GABAergic hypoalgesic interneurons exist in the dorsal horn in neuropathic pain model animals, the recovery of impaired GABAergic inhibition in the dorsal horn may improve pain behavior. We herein determined whether the production of gamma-aminobutyric acid (GABA) and glutamic acid decarboxylase (GAD) in the dorsal horn is restored by treadmill running and contributes to exercise-induced hypoalgesia in neuropathic pain model mice. C57BL/6 J mice underwent partial sciatic nerve ligation (PSL). PSL-Runner mice ran on a treadmill at 7 m/min for 60 min/day, 5 days/week, from two days after PSL. Results Mechanical allodynia and heat hyperalgesia developed in PSL-Sedentary mice but were significantly attenuated in PSL-Runner mice. PSL markedly decreased GABA and GAD65/67 levels in neuropils in the ipsilateral dorsal horn, while treadmill running inhibited these reductions. GABA+ neuronal nuclei+ interneuron numbers in the ipsilateral dorsal horn were significantly decreased in PSL-Sedentary mice but not in PSL-Runner mice. Pain behavior thresholds positively correlated with GABA and GAD65/67 levels and GABAergic interneuron numbers in the ipsilateral dorsal horns of PSL-Sedentary and -Runner mice. Conclusions Treadmill running prevented PSL-induced reductions in GAD65/67 production, and, thus, GABA levels may be retained in interneurons and neuropils in the superficial dorsal horn. Therefore, improvements in impaired GABAergic inhibition may be involved in exercise-induced hypoalgesia.
Collapse
Affiliation(s)
- Katsuya Kami
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Satoru Taguchi Ms
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Fumihiro Tajima
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Emiko Senba
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan Department of Physical Therapy, Osaka Yukioka College of Health Science, Ibaraki City, Osaka, Japan
| |
Collapse
|
21
|
Abstract
Among many mechanisms implicated in the development of neuropathic pain after nerve damage is a profound dysfunction of GABAergic inhibitory controls, manifested by ongoing pain, mechanical hypersensitivity, and thermal hyperalgesia. In some respects, neuropathic pain can be considered a "disease" of the nervous system, with features in common with trauma-induced seizures. Indeed, first-line management involves anticonvulsant therapy. An alternative to pharmacotherapy for neuropathic pain is an approach that reestablishes the inhibitory tone that is lost after nerve damage. To this end, we have transplanted embryonic cortical GABAergic precursor neurons into the spinal cord of nerve-injured mice. Using a combination of light and electron microscopic analyses, and also in vitro electrophysiological recordings from spinal cord slice preparations, we demonstrated remarkable integration of the transplants into the host, adult spinal cord. Most importantly, transplants produced a complete reversal of the hypersensitivity in a sciatic nerve injury model and in a paclitaxel-generated chemotherapy model of neuropathic pain. In related studies, we demonstrated that medial ganglionic eminence cell transplants are also effective in a chronic neuropathic itch model in which there is a significant loss of dorsal horn inhibitory interneurons. Most importantly, in contrast to systemic or intrathecal pharmacological therapies, adverse side effects are minimized when the inhibitory control, namely, γ-aminobutyric acid release, occurs in a spinal cord circuit. These studies suggest that therapy targeted at repairing the GABAergic dysfunction is a viable and novel alternative to the management of neuropathic pain and itch, particularly those that are or become refractory to traditional pharmacotherapy.
Collapse
Affiliation(s)
- Allan I Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
22
|
Hanani M. Role of satellite glial cells in gastrointestinal pain. Front Cell Neurosci 2015; 9:412. [PMID: 26528140 PMCID: PMC4602093 DOI: 10.3389/fncel.2015.00412] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 09/28/2015] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal (GI) pain is a common clinical problem, for which effective therapy is quite limited. Sensations from the GI tract, including pain, are mediated largely by neurons in the dorsal root ganglia (DRG), and to a smaller extent by vagal afferents emerging from neurons in the nodose/jugular ganglia. Neurons in rodent DRG become hyperexcitable in models of GI pain (e.g., gastric or colonic inflammation), and can serve as a source for chronic pain. Glial cells are another element in the pain signaling pathways, and there is evidence that spinal glial cells (microglia and astrocytes) undergo activation (gliosis) in various pain models and contribute to pain. Recently it was found that satellite glial cells (SGCs), the main type of glial cells in sensory ganglia, might also contribute to chronic pain in rodent models. Most of that work focused on somatic pain, but in several studies GI pain was also investigated, and these are discussed in the present review. We have shown that colonic inflammation induced by dinitrobenzene sulfonic acid (DNBS) in mice leads to the activation of SGCs in DRG and increases gap junction-mediated coupling among these cells. This coupling appears to contribute to the hyperexcitability of DRG neurons that innervate the colon. Blocking gap junctions (GJ) in vitro reduced neuronal hyperexcitability induced by inflammation, suggesting that glial GJ participate in SGC-neuron interactions. Moreover, blocking GJ by carbenoxolone and other agents reduces pain behavior. Similar changes in SGCs were also found in the mouse nodose ganglia (NG), which provide sensory innervation to most of the GI tract. Following systemic inflammation, SGCs in these ganglia were activated, and displayed augmented coupling and greater sensitivity to the pain mediator ATP. The contribution of these changes to visceral pain remains to be determined. These results indicate that although visceral pain is unique, it shares basic mechanisms with somatic pain, suggesting that therapeutic approaches to both pain types may be similar. Future research in this field should include additional types of GI injury and also other types of visceral pain.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus Jerusalem, Israel
| |
Collapse
|
23
|
Ford A, Castonguay A, Cottet M, Little JW, Chen Z, Symons-Liguori AM, Doyle T, Egan TM, Vanderah TW, De Koninck Y, Tosh DK, Jacobson KA, Salvemini D. Engagement of the GABA to KCC2 signaling pathway contributes to the analgesic effects of A3AR agonists in neuropathic pain. J Neurosci 2015; 35:6057-67. [PMID: 25878279 PMCID: PMC4397603 DOI: 10.1523/jneurosci.4495-14.2015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 01/18/2023] Open
Abstract
More than 1.5 billion people worldwide suffer from chronic pain, yet current treatment strategies often lack efficacy or have deleterious side effects in patients. Adenosine is an inhibitory neuromodulator that was previously thought to mediate antinociception through the A1 and A2A receptor subtypes. We have since demonstrated that A3AR agonists have potent analgesic actions in preclinical rodent models of neuropathic pain and that A3AR analgesia is independent of adenosine A1 or A2A unwanted effects. Herein, we explored the contribution of the GABA inhibitory system to A3AR-mediated analgesia using well-characterized mouse and rat models of chronic constriction injury (CCI)-induced neuropathic pain. The deregulation of GABA signaling in pathophysiological pain states is well established: GABA signaling can be hampered by a reduction in extracellular GABA synthesis by GAD65 and enhanced extracellular GABA reuptake via the GABA transporter, GAT-1. In neuropathic pain, GABAAR-mediated signaling can be further disrupted by the loss of the KCC2 chloride anion gradient. Here, we demonstrate that A3AR agonists (IB-MECA and MRS5698) reverse neuropathic pain via a spinal mechanism of action that modulates GABA activity. Spinal administration of the GABAA antagonist, bicuculline, disrupted A3AR-mediated analgesia. Furthermore, A3AR-mediated analgesia was associated with reductions in CCI-related GAD65 and GAT-1 serine dephosphorylation as well as an enhancement of KCC2 serine phosphorylation and activity. Our results suggest that A3AR-mediated reversal of neuropathic pain increases modulation of GABA inhibitory neurotransmission both directly and indirectly through protection of KCC2 function, underscoring the unique utility of A3AR agonists in chronic pain.
Collapse
Affiliation(s)
- Amanda Ford
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Annie Castonguay
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Martin Cottet
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Joshua W Little
- Department of Surgery, Center for Anatomical Science and Education, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Zhoumou Chen
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Ashley M Symons-Liguori
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724-5050, and
| | - Timothy Doyle
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Terrance M Egan
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Todd W Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724-5050, and
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810
| | - Daniela Salvemini
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104,
| |
Collapse
|
24
|
Kami K, Taguchi S, Tajima F, Senba E. Mechanisms and effects of forced and voluntary exercises on exercise-induced hypoalgesia in neuropathic pain model mice. ACTA ACUST UNITED AC 2015. [DOI: 10.11154/pain.30.216] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Katsuya Kami
- Department of Rehabilitation Medicine, Wakayama Medical University
| | - Satoru Taguchi
- Department of Rehabilitation Medicine, Wakayama Medical University
| | - Fumihiro Tajima
- Department of Rehabilitation Medicine, Wakayama Medical University
| | - Emiko Senba
- Department of Physical Therapy, Osaka Yukioka College of Health Science
| |
Collapse
|
25
|
Activation of GABA(B) receptors potentiates inward rectifying potassium currents in satellite glial cells from rat trigeminal ganglia: in vivo patch-clamp analysis. Neuroscience 2014; 288:51-8. [PMID: 25542421 DOI: 10.1016/j.neuroscience.2014.12.024] [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: 11/12/2014] [Revised: 12/15/2014] [Accepted: 12/15/2014] [Indexed: 11/23/2022]
Abstract
In a previous study, we demonstrated that inflammation suppressed inward rectifying K(+) (Kir) currents in satellite glial cells (SGCs) from the trigeminal ganglia (TRGs) and that this impairment of glial potassium homeostasis in the trigeminal ganglion (TRG) contributed to trigeminal pain. The aim of the present study was to investigate whether activation of GABAB receptors modulates the Kir current in SGCs using in vivo patch-clamp and immunohistochemical techniques. Immunohistochemically, we found that immunoreactivity for glial-specific Kir channel subunit Kir4.1 and the GABAB receptor was co-expressed in SGCs from the TRGs. In vivo whole-cell recordings were made using SGCs from the TRGs of urethane-anesthetized rats. Application of baclofen, a GABAB receptor agonist, significantly increased the mean peak amplitude of Kir currents in a concentration-dependent and reversible manner. Baclofen-induced potentiation of the Kir current was abolished by co-application of 3-amino-2-(4-chlorophenyl)-2-hydroxyprophylsulfonic acid (saclofen). In addition, baclofen significantly potentiated the density of the Ba(2+)-sensitive Kir current, and resulted in hyperpolarization of the mean membrane potential. These results suggest that activation of GABAB receptors potentiates the Kir current in SGCs and that GABA released from the TRG neuronal soma could contribute to buffering of extracellular K(+) concentrations following excitation of TRG neurons during the processing of sensory information, including the transmission of noxious stimuli.
Collapse
|
26
|
Chen X, Wang T, Lin C, Chen B. Effect of adenoviral delivery of prodynorphin gene on experimental inflammatory pain induced by formalin in rats. Int J Clin Exp Med 2014; 7:4877-4886. [PMID: 25663984 PMCID: PMC4307431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 11/13/2014] [Indexed: 06/04/2023]
Abstract
Circumstantial evidences suggest that dynorphins and their common precursor prodynorphin (PDYN) are involved in antinociception and neuroendocrine signaling. DREAM knockout mice had increased levels of PDYN and dynorphin expression, and reduced sensitivity to painful stimuli. However, some data support the notion that the up-regulation of spinal dynorphin expression is a common critical feature in neuropathic pain. It is not clear whether the production of dynorphin A can be increased when more PDYN is present. In this study we investigated the changes in pain behaviors, spinal PDYN mRNA expression and dynorphin A production on formalin-induced pain in rats receiving the pretreatment of adenoviral delivery of PDYN. Our results showed that the adenoviral transfer of PDYN gene was sufficient to reduce pain behaviors resulting from formalin injection, and the antinociceptive effect after receiving the pretreatment of adenoviral delivery of PDYN was mediated at the level of the spinal cord via KOR.
Collapse
Affiliation(s)
- Xionggang Chen
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350005, P. R. China
| | - Tingting Wang
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350005, P. R. China
| | - Caizhu Lin
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350005, P. R. China
| | - Baihong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical UniversityFuzhou 350005, P. R. China
| |
Collapse
|
27
|
Yang ES, Bae JY, Kim TH, Kim YS, Suk K, Bae YC. Involvement of endoplasmic reticulum stress response in orofacial inflammatory pain. Exp Neurobiol 2014; 23:372-80. [PMID: 25548537 PMCID: PMC4276808 DOI: 10.5607/en.2014.23.4.372] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/22/2014] [Accepted: 10/22/2014] [Indexed: 01/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is involved in many neurological diseases and inflammatory responses. Inflammatory mediators induce neuronal damage and trigger the neuropathic or inflammatory pain. But there is very little data on the role of the ER stress response in pain mechanisms. In this study, we explored whether the ER stress response is involved in orofacial inflammatory pain by using a complete Freund's adjuvant (CFA)-injected rat model. The thermal pain hypersensitivity increased significantly after CFA injection. We found that the protein and mRNA levels of ER stress response genes, GRP78/Bip and p-eIF2α, increased significantly in trigeminal ganglion (TG) of CFA-injected rats compared to control animals. In immunofluorescence analysis, a significant increase of GRP78 and p-eIF2α immunopositive neurons was observed in CFA-injected TG compared to control TG. When we administered an ER stress modulator, salubrinal, CFA-induced thermal pain hypersensitivity was temporally reduced. Thus, our study suggests that ER stress responses in TG neurons contribute to CFA-induced inflammatory pain, and may comprise an important molecular mechanism underlying the orofacial inflammatory pain pathway.
Collapse
Affiliation(s)
- Eun Sun Yang
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Jin Young Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Tae Heon Kim
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Yun Sook Kim
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| |
Collapse
|
28
|
Kramer PR, Bellinger LL. Infusion of Gabrα6 siRNA into the trigeminal ganglia increased the myogenic orofacial nociceptive response of ovariectomized rats treated with 17β-estradiol. Neuroscience 2014; 278:144-53. [PMID: 25128322 PMCID: PMC4172543 DOI: 10.1016/j.neuroscience.2014.07.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 01/06/2023]
Abstract
High levels of 17β-estradiol (E2) have been found to reduce inflammatory temporomandibular joint (TMJ) pain. A search for genes effected by a high concentration of estradiol showed an increase in GABAA receptor subunit alpha 6 (Gabrα6) in the trigeminal ganglia (TG). Blockade of Gabrα6 expression in the TG increases masseter muscle nociception in male rats, but the relationship between estradiol's effect on nociception and Gabrα6 expression remains unclear in females. To address this knowledge gap we hypothesized that reducing Gabrα6 expression in the TG will increase the orofacial nociceptive response of ovariectomized female rats treated with estradiol. To administer hormone osmotic pumps were placed in rats that dispensed a low diestrus plasma concentration of 17β-estradiol, in addition, 17β-estradiol was injected to produce a high proestrus plasma concentration of estradiol. A ligature was then placed around the masseter tendon to induce a nociceptive response; a model for TMJ muscle pain. Gabrα6 small interfering RNA (siRNA) was later infused into the TG and the nociceptive response was measured using von Frey filaments and a meal duration assay. GABAA receptor expression was measured in the TG and trigeminal nucleus caudalis and upper cervical region (Vc-C1). Ligature significantly increased the nociceptive response but a high proestrus concentration of 17β-estradiol attenuated this response. Gabrα6 siRNA infusion decreased Gabrα6 expression in the TG and Vc-C1 but increased the nociceptive response after 17β-estradiol treatment. The results suggest estradiol decreased the orofacial nociceptive response, in part, by causing an increase in Gabrα6 expression.
Collapse
Affiliation(s)
- P R Kramer
- Department of Biomedical Sciences, Texas A&M University, Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States.
| | - L L Bellinger
- Department of Biomedical Sciences, Texas A&M University, Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States
| |
Collapse
|
29
|
Sokolov AY, Lyubashina OA, Amelin AV, Panteleev SS. The role of gamma-aminobutyric acid in migraine pathogenesis. NEUROCHEM J+ 2014. [DOI: 10.1134/s1819712414020093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
30
|
Tzabazis AZ, Klukinov M, Feliciano DP, Wilson SP, Yeomans DC. Gene therapy for trigeminal pain in mice. Gene Ther 2014; 21:422-6. [PMID: 24572785 PMCID: PMC3975690 DOI: 10.1038/gt.2014.14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 12/09/2013] [Accepted: 01/06/2014] [Indexed: 12/11/2022]
Abstract
The aim of this study was to test the efficacy of a single direct injection of viral vector encoding for encephalin to induce a widespread expression of the transgene and potential analgesic effect in trigeminal behavioral pain models in mice. After direct injection of HSV-1 based vectors encoding for human preproenkephalin (SHPE) or the lacZ reporter gene (SHZ.1, control virus) into the trigeminal ganglia in mice, we performed an orofacial formalin test and assessed the cumulative nociceptive behavior at different time points after injection of the viral vectors. We observed an analgesic effect on nociceptive behavior that lasted up to 8 weeks after a single injection of SHPE into the trigeminal ganglia. Control virus injected animals showed nociceptive behavior similar to naïve mice. The analgesic effect of SHPE injection was reversed/attenuated by subcutaneous naloxone injections, a μ-opioid receptor antagonist. SHPE injected mice also showed normalization in withdrawal latencies upon thermal noxious stimulation of inflamed ears after subdermal complete Freund’s adjuvans injection indicating widespread expression of the transgene. Quantitative immunohistochemistry of trigeminal ganglia showed expression of human preproenkephalin after SHPE injection. Direct injection of viral vectors proved to be useful for exploring the distinct pathophysiology of the trigeminal system and could also be an interesting addition to the pain therapists’ armamentarium.
Collapse
Affiliation(s)
- A Z Tzabazis
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - M Klukinov
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - D P Feliciano
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - S P Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - D C Yeomans
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
31
|
Kramer PR, Bellinger LL. Reduced GABAA receptor α6 expression in the trigeminal ganglion enhanced myofascial nociceptive response. Neuroscience 2013; 245:1-11. [PMID: 23602886 DOI: 10.1016/j.neuroscience.2013.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 04/01/2013] [Accepted: 04/02/2013] [Indexed: 10/26/2022]
Abstract
Activation of the GABAA receptor results in inhibition of neuronal activity. One subunit of this multi-subunit receptor termed alpha 6 (Gabrα6) contributed to inflammatory temporomandibular joint (TMJ) nociception but TMJ disorders often include myofascial pain. To address Gabrα6 role in myofascial pain we hypothesized that Gabrα6 has an inhibitory role in myofascial nociceptive responses similar to inflammatory TMJ arthritis. To test this hypothesis a, myofascial nociceptive response was induced by placing a ligature bilaterally on the tendon attachment of the anterior superficial part of a male rat's masseter muscle. Four days after ligature placement Gabrα6 expression was reduced by infusing the trigeminal ganglia (TG) with small interfering RNA (siRNA) having homology to either the Gabrα6 gene (Gabrα6 siRNA) or no known gene (control siRNA). After siRNA infusion nociceptive behavioral responses were measured, i.e., feeding behavior and head withdrawal after pressing upon the region above the ligature with von Frey filaments. Neuronal activity in the TG and trigeminal nucleus caudalis and upper cervical region (Vc-C1) was measured by quantitating the amount of phosphorylated extracellular signal-regulated kinase (p-ERK). Total Gabrα6 and GABAA receptor contents in the TG and Vc-C1 were determined. Gabrα6 siRNA infusion reduced Gabrα6 and GABAA receptor expression and significantly increased the nociceptive response in both nociceptive assays. Gabrα6 siRNA infusion also significantly increased TG p-ERK expression of the ligated rats. From these results we conclude GABAA receptors consisting of the Gabrα6 subunit inhibit TG nociceptive sensory afferents in the trigeminal pathway and have an important role in the regulation of myofascial nociception.
Collapse
Affiliation(s)
- P R Kramer
- Department of Biomedical Sciences, Texas A&M Health Science Center Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States.
| | | |
Collapse
|
32
|
Bráz JM, Sharif-Naeini R, Vogt D, Kriegstein A, Alvarez-Buylla A, Rubenstein JL, Basbaum AI. Forebrain GABAergic neuron precursors integrate into adult spinal cord and reduce injury-induced neuropathic pain. Neuron 2012; 74:663-75. [PMID: 22632725 DOI: 10.1016/j.neuron.2012.02.033] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2012] [Indexed: 10/28/2022]
Abstract
Neuropathic pain is a chronic debilitating disease characterized by mechanical allodynia and spontaneous pain. Because symptoms are often unresponsive to conventional methods of pain treatment, new therapeutic approaches are essential. Here, we describe a strategy that not only ameliorates symptoms of neuropathic pain but is also potentially disease modifying. We show that transplantation of immature telencephalic GABAergic interneurons from the mouse medial ganglionic eminence (MGE) into the adult mouse spinal cord completely reverses the mechanical hypersensitivity produced by peripheral nerve injury. Underlying this improvement is a remarkable integration of the MGE transplants into the host spinal cord circuitry, in which the transplanted cells make functional connections with both primary afferent and spinal cord neurons. By contrast, MGE transplants were not effective against inflammatory pain. Our findings suggest that MGE-derived GABAergic interneurons overcome the spinal cord hyperexcitability that is a hallmark of nerve injury-induced neuropathic pain.
Collapse
Affiliation(s)
- João M Bráz
- Department of Anatomy, W.M. Keck Foundation Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Heterogenous GABAB receptor-mediated pathways are involved in the local GABAergic system of the rat trigeminal ganglion: Possible involvement of KCTD proteins. Neuroscience 2012; 218:344-58. [DOI: 10.1016/j.neuroscience.2012.05.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 05/01/2012] [Accepted: 05/14/2012] [Indexed: 01/28/2023]
|
34
|
Goins WF, Cohen JB, Glorioso JC. Gene therapy for the treatment of chronic peripheral nervous system pain. Neurobiol Dis 2012; 48:255-70. [PMID: 22668775 DOI: 10.1016/j.nbd.2012.05.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 05/11/2012] [Accepted: 05/24/2012] [Indexed: 11/30/2022] Open
Abstract
Chronic pain is a major health concern affecting 80 million Americans at some time in their lives with significant associated morbidity and effects on individual quality of life. Chronic pain can result from a variety of inflammatory and nerve damaging events that include cancer, infectious diseases, autoimmune-related syndromes and surgery. Current pharmacotherapies have not provided an effective long-term solution as they are limited by drug tolerance and potential abuse. These concerns have led to the development and testing of gene therapy approaches to treat chronic pain. The potential efficacy of gene therapy for pain has been reported in numerous pre-clinical studies that demonstrate pain control at the level of the spinal cord. This promise has been recently supported by a Phase-I human trial in which a replication-defective herpes simplex virus (HSV) vector was used to deliver the human pre-proenkephalin (hPPE) gene, encoding the natural opioid peptides met- and leu-enkephalin (ENK), to cancer patients with intractable pain resulting from bone metastases (Fink et al., 2011). The study showed that the therapy was well tolerated and that patients receiving the higher doses of therapeutic vector experienced a substantial reduction in their overall pain scores for up to a month post vector injection. These exciting early clinical results await further patient testing to demonstrate treatment efficacy and will likely pave the way for other gene therapies to treat chronic pain.
Collapse
Affiliation(s)
- William F Goins
- Dept of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh PA 15219, USA.
| | | | | |
Collapse
|
35
|
Puri J, Vinothini P, Reuben J, Bellinger LL, Ailing L, Peng YB, Kramer PR. Reduced GABA(A) receptor α6 expression in the trigeminal ganglion alters inflammatory TMJ hypersensitivity. Neuroscience 2012; 213:179-90. [PMID: 22521829 DOI: 10.1016/j.neuroscience.2012.03.059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 03/15/2012] [Accepted: 03/16/2012] [Indexed: 11/28/2022]
Abstract
Trigeminal ganglia neurons express the GABA(A) receptor subunit alpha 6 (Gabrα6) but the role of this particular subunit in orofacial hypersensitivity is unknown. In this report the function of Gabrα6 was tested by reducing its expression in the trigeminal ganglia and measuring the effect of this reduction on inflammatory temporomandibular joint (TMJ) hypersensitivity. Gabrα6 expression was reduced by infusing the trigeminal ganglia of male Sprague Dawley rats with small interfering RNA (siRNA) having homology to either the Gabrα6 gene (Gabrα6 siRNA) or no known gene (control siRNA). Sixty hours after siRNA infusion the rats received a bilateral TMJ injection of complete Freund's adjuvant to induce an inflammatory response. Hypersensitivity was then quantitated by measuring meal duration, which lengthens when hypersensitivity increases. Neuronal activity in the trigeminal ganglia was also measured by quantitating the amount of phosphorylated ERK. Rats in a different group that did not have TMJ inflammation had an electrode placed in the spinal cord at the level of C1 sixty hours after siRNA infusion to record extracellular electrical activity of neurons that responded to TMJ stimulation. Our results show that Gabrα6 was expressed in both neurons and satellite glia of the trigeminal ganglia and that Gabrα6 positive neurons within the trigeminal ganglia have afferents in the TMJ. Gabrα6 siRNA infusion reduced Gabrα6 gene expression by 30% and significantly lengthened meal duration in rats with TMJ inflammation. Gabrα6 siRNA infusion also significantly increased p-ERK expression in the trigeminal ganglia of rats with TMJ inflammation and increased electrical activity in the spinal cord of rats without TMJ inflammation. These results suggest that maintaining Gabrα6 expression was necessary to inhibit primary sensory afferents in the trigeminal pathway and reduce inflammatory orofacial nociception.
Collapse
Affiliation(s)
- J Puri
- Department of Biomedical Sciences, Texas A&M Health Science Center, Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Gad2 encodes GAD65, which is present preferentially in presynaptic terminals for synthesis of GABA for vesicle release. Gad2 is a regulatory target of cell activities in various brain functions and in GABA perturbation-related neurological diseases. However, our understanding of how Gad2 is transcriptionally regulated and how Gad2 transcription responds to changing cell environment under these conditions is still limited. This review discusses recent advances in the regulatory mechanisms for Gad2 transcription and highlights the characteristics of TATA-less Gad2 promoters and regulation of Gad2 transcription by CREB and by activity-dependent epigenetic modification of the chromatin structure in regulatory elements of the Gad2 gene.
Collapse
Affiliation(s)
- Zhizhong Z Pan
- Department of Anesthesiology and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
37
|
Lee KY, Charbonnet M, Gold MS. Upregulation of high-affinity GABA(A) receptors in cultured rat dorsal root ganglion neurons. Neuroscience 2012; 208:133-42. [PMID: 22366297 DOI: 10.1016/j.neuroscience.2012.01.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 01/27/2012] [Accepted: 01/31/2012] [Indexed: 10/14/2022]
Abstract
Despite evidence that high-affinity GABA(A) receptor subunit mRNA and protein are present in dorsal root ganglia (DRG), low-affinity currents dominate those detected in acutely dissociated DRG neurons in vitro. This observation raises the possibility that high-affinity receptors are normally trafficked out of the DRG toward central and peripheral terminals. We therefore hypothesized that with time in culture, there would be an increase in high-affinity GABA(A) currents in DRG neurons. To test this hypothesis, we studied dissociated DRG neurons 2 h (acute) and 24 h (cultured) after plating with whole-cell patch-clamp techniques, Western blot, and semiquantitative reverse transcriptase polymerase chain reaction (sqRT-PCR) analysis. GABA(A) current density increases dramatically with time in culture in association with the emergence of two persistent currents with EC50's of 0.25±0.01 μM and 3.2±0.02 μM for GABA activation. In a subpopulation of neurons, there was also an increase in the potency of GABA activation of the transient current from an EC50 of 78.16±10.1 μM to 9.56±1.3 μM with time in culture. A fraction of the high-affinity current was potentiated by δ-subunit agonist 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridine-3-ol (THIP). δ-subunit immunoreactivity was largely restricted to the cytosolic fraction in acute, but the membrane fraction in cultured, DRG neurons, with no detectable change in δ-subunit mRNA. However, the emergence of a high-affinity current blocked by THIP and insensitive to bicuculline was detected in a subpopulation of cultured neurons as well in association with an increase in ρ2- and ρ3-subunit mRNA in cultured DRG neurons. Our results suggest that high-affinity δ-subunit-containing GABA(A) receptors are normally trafficked out of the DRG where they are targeted to peripheral and central processes. They also highlight that the interpretation of data obtained from cultured DRG neurons should be made with caution.
Collapse
Affiliation(s)
- K Y Lee
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | |
Collapse
|
38
|
Abstract
Orofacial pain remains an understudied area in pain research given that most attention has been focused on the spinal system. In this chapter, animal models of neuropathic and inflammatory orofacial pain are presented. Four different types of pain behavior tests are then described for assessing evoked and spontaneous pain behavior in addition to conditional reward behavior. The use of a combination of different pain models and behavior assessments is needed to aid in understanding the mechanisms contributing to orofacial pain in humans for developing effective therapy.
Collapse
Affiliation(s)
- Timothy K Y Kaan
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
39
|
Zhang Z, Cai YQ, Zou F, Bie B, Pan ZZ. Epigenetic suppression of GAD65 expression mediates persistent pain. Nat Med 2011; 17:1448-55. [PMID: 21983856 PMCID: PMC3210928 DOI: 10.1038/nm.2442] [Citation(s) in RCA: 210] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 07/12/2011] [Indexed: 12/19/2022]
Abstract
Chronic pain is a common neurological disease involving lasting, multifaceted maladaptations ranging from gene modulation to synaptic dysfunction and emotional disorders. Sustained pathological stimuli in many diseases alter the output activities of certain genes through epigenetic modifications, but it is unclear how epigenetic mechanisms operate in the development of chronic pain. We show here that in the rat brainstem nucleus raphe magnus, which is important for central mechanisms of chronic pain, persistent inflammatory and neuropathic pain epigenetically suppresses Gad2 (encoding glutamic acid decarboxylase 65 (GAD65)) transcription through histone deacetylase (HDAC)-mediated histone hypoacetylation, resulting in impaired γ-aminobutyric acid (GABA) synaptic inhibition. Gad2 knockout mice showed sensitized pain behavior and impaired GABA synaptic function in their brainstem neurons. In wild-type but not Gad2 knockout mice, HDAC inhibitors strongly increased GAD65 activity, restored GABA synaptic function and relieved sensitized pain behavior. These findings suggest GAD65 and HDACs as potential therapeutic targets in an epigenetic approach to the treatment of chronic pain.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | | | | | | | | |
Collapse
|
40
|
Handy CR, Krudy C, Boulis N. Gene therapy: a potential approach for cancer pain. PAIN RESEARCH AND TREATMENT 2011; 2011:987597. [PMID: 22110939 PMCID: PMC3196247 DOI: 10.1155/2011/987597] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 12/14/2010] [Accepted: 01/21/2011] [Indexed: 12/21/2022]
Abstract
Chronic pain is experienced by as many as 90% of cancer patients at some point during the disease. This pain can be directly cancer related or arise from a sensory neuropathy related to chemotherapy. Major pharmacological agents used to treat cancer pain often lack anatomical specificity and can have off-target effects that create new sources of suffering. These concerns establish a need for improved cancer pain management. Gene therapy is emerging as an exciting prospect. This paper discusses the potential for viral vector-based treatment of cancer pain. It describes studies involving vector delivery of transgenes to laboratory pain models to modulate the nociceptive cascade. It also discusses clinical investigations aimed at regulating pain in cancer patients. Considering the prevalence of pain among cancer patients and the growing potential of gene therapy, these studies could set the stage for a new class of medicines that selectively disrupt nociceptive signaling with limited off-target effects.
Collapse
Affiliation(s)
- Chalonda R. Handy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| | - Christina Krudy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| |
Collapse
|
41
|
|
42
|
Handy CR, Krudy C, Boulis N, Federici T. Pain in amyotrophic lateral sclerosis: a neglected aspect of disease. Neurol Res Int 2011; 2011:403808. [PMID: 21766021 PMCID: PMC3135011 DOI: 10.1155/2011/403808] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 03/06/2011] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder marked by progressive loss of motor neurons, muscle wasting, and respiratory dysfunction. With disease progression, secondary symptoms arise creating new problematic conditions for ALS patients. Amongst these is pain. Although not a primary consequence of disease, pain occurs in a substantial number of individuals. Yet, studies investigating its pathomechanistic properties in the ALS patient are lacking. Therefore, more exploratory efforts into its scope, severity, impact, and treatment should be initiated. Several studies investigating the use of Clostridial neurotoxins for the reduction of pain in ALS patients suggest the potential for a neural specific approach involving focal drug delivery. Gene therapy represents a way to accomplish this. Therefore, the use of viral vectors to express transgenes that modulate the nociceptive cascade could prove to be an effective way to achieve meaningful benefit in conditions of pain in ALS.
Collapse
Affiliation(s)
- Chalonda R. Handy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA
| | - Christina Krudy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA
| | - Thais Federici
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA
| |
Collapse
|
43
|
Huang Y, Liu X, Dong L, Liu Z, He X, Liu W. Development of viral vectors for gene therapy for chronic pain. PAIN RESEARCH AND TREATMENT 2011; 2011:968218. [PMID: 22110937 PMCID: PMC3200086 DOI: 10.1155/2011/968218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 01/31/2011] [Indexed: 11/17/2022]
Abstract
Chronic pain is a major health concern that affects millions of people. There are no adequate long-term therapies for chronic pain sufferers, leading to significant cost for both society and the individual. The most commonly used therapy for chronic pain is the application of opioid analgesics and nonsteroidal anti-inflammatory drugs, but these drugs can lead to addiction and may cause side effects. Further studies of the mechanisms of chronic pain have opened the way for development of new treatment strategies, one of which is gene therapy. The key to gene therapy is selecting safe and highly efficient gene delivery systems that can deliver therapeutic genes to overexpress or suppress relevant targets in specific cell types. Here we review several promising viral vectors that could be applied in gene transfer for the treatment of chronic pain and further discuss the possible mechanisms of genes of interest that could be delivered with viral vectors for the treatment of chronic pain.
Collapse
Affiliation(s)
- Yu Huang
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Xin Liu
- College of Pharmacy, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Lanlan Dong
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Zhongchun Liu
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Xiaohua He
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
- Research Center of Food and Drug Evaluation, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| | - Wanhong Liu
- School of Medicine, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
- Research Center of Food and Drug Evaluation, Wuhan University, Donghu Road #185, Wuchang, Wuhan 430071, China
| |
Collapse
|
44
|
Abstract
Satellite glial cells (SGCs) undergo phenotypic changes and divide the following injury into a peripheral nerve. Nerve injury, also elicits an immune response and several antigen-presenting cells are found in close proximity to SGCs. Silencing SCG-specific molecules involved in intercellular transport (Connexin 43) or glutamate recycling (glutamine synthase) can dramatically alter nociceptive responses of normal and nerve-injured rats. Transducing SGCs with glutamic acid decarboxylase can produce analgesia in models of trigeminal pain. Taken together these data suggest that SGCs may play a role in the genesis or maintenance of pain and open a range of new possibilities for curing neuropathic pain.
Collapse
|