101
|
Shah RM, Peterson C, Strom A, Dillenburg M, Finzel B, Kitto KF, Fairbanks C, Wilcox G, Wagner CR. Inhibition of HINT1 Modulates Spinal Nociception and NMDA Evoked Behavior in Mice. ACS Chem Neurosci 2019; 10:4385-4393. [PMID: 31503445 DOI: 10.1021/acschemneuro.9b00432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The interactions between the mu-opioid (MOR) and N-methyl-d-aspartate receptor (NMDAR) constitute an area of intense investigation because of their contributions to maladaptive neuroplasticity. Recent evidence suggests that their association requires the involvement of histidine triad nucleotide-binding protein (HINT1) with the enzyme's active site being critical in its regulatory role. Since it is known that spinal blockade of NMDA receptors prevents the development of opioid analgesic tolerance, we hypothesized that spinal inhibition of the HINT1 enzyme may similarly inhibit opioid tolerance. To address these questions, we evaluated novel HINT1 active-site inhibitors in two models of NMDAR and MOR interaction, namely, MOR inhibition of spinal NMDA activation and acute endomorphin-2 tolerance. These studies revealed that while the tryptamine carbamate of guanosine inhibitor, TrpGc, blocked both the development of opioid tolerance and the inhibitory effect of opioids on NMDA activation of the NMDA receptor, acyl-sulfamate analogues could only block the latter. Thermodynamic binding and X-ray crystallographic studies suggested that there are key differences between the bound HINT1-inhibitor surfaces that may be responsible for their differential ability to probe the ability of HINT1 to regulate cross talk between the mu-opioid receptor and NMDA receptor in the spinal cord.
Collapse
|
102
|
Lesnak J, Sluka KA. Chronic non-inflammatory muscle pain: central and peripheral mediators. CURRENT OPINION IN PHYSIOLOGY 2019; 11:67-74. [PMID: 31998857 PMCID: PMC6988739 DOI: 10.1016/j.cophys.2019.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Conditions with chronic widespread non-inflammatory muscle pain, such as fibromyalgia, have complex etiologies with numerous proposed mechanisms for their pathophysiology of underlying chronic pain. Advancements in neuroimaging have allowed for the study of brain function and connectivity in humans with these conditions, while development of animal models have allowed for the study of both peripheral and central factors that lead to chronic pain. This article reviews the current literature surrounding the pathophysiology of chronic widespread non-inflammatory muscle pain focusing on both peripheral and central nervous system, as well as immune system, contributions to the development and maintenance of pain. A better understanding of the mechanisms underlying these conditions can allow for improvements in patient education, treatment and outcomes.
Collapse
Affiliation(s)
- Joseph Lesnak
- Department of Physical Therapy and Rehabilitation Science, Pain Research Program, 1-242 MEB, University of Iowa, Iowa City, IA 52252, USA
| | - Kathleen A. Sluka
- Department of Physical Therapy and Rehabilitation Science, Pain Research Program, 1-242 MEB, University of Iowa, Iowa City, IA 52252, USA
| |
Collapse
|
103
|
Schreiber KL, Belfer I, Miaskowski C, Schumacher M, Stacey BR, Van De Ven T. AAAPT Diagnostic Criteria for Acute Pain Following Breast Surgery. THE JOURNAL OF PAIN 2019; 21:294-305. [PMID: 31493489 DOI: 10.1016/j.jpain.2019.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/30/2022]
Abstract
Acute pain after breast surgery decreases the quality of life of cancer survivors. Previous studies using a variety of definitions and methods report prevalence rates between 10% and 80%, which suggests the need for a comprehensive framework that can be used to guide assessment of acute pain and pain-related outcomes after breast surgery. A multidisciplinary task force with clinical and research expertise performed a focused review and synthesis and applied the 5 dimensional framework of the AAAPT (Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks [ACTTION], American Academy of Pain Medicine [AAPM], American Pain Society [APS] Pain Taxonomy) to acute pain after breast surgery. Application of the AAAPT taxonomy yielded the following: 1) Core Criteria: Location, timing, severity, and impact of breast surgery pain were defined; 2) Common Features: Character and expected trajectories were established in relevant surgical subgroups, and common pain assessment tools for acute breast surgery pain identified; 3) Modulating Factors: Biological, psychological, and social factors that modulate interindividual variability were delineated; 4) Impact/Functional Consequences: Domains of impact were outlined and defined; 5) Neurobiologic Mechanisms: Putative mechanisms were specified ranging from nerve injury, inflammation, peripheral and central sensitization, to affective and social processing of pain. PERSPECTIVE: The AAAPT provides a framework to define and guide improved assessment of acute pain after breast surgery, which will enhance generalizability of results across studies and facilitate meta-analyses and studies of interindividual variation, and underlying mechanism. It will allow researchers and clinicians to better compare between treatments, across institutions, and with other types of acute pain.
Collapse
Affiliation(s)
- Kristin L Schreiber
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Inna Belfer
- National Center for Complementary and Integrative Health, NIH, Bethesda, Maryland
| | - Christine Miaskowski
- Department of Physiological Nursing, University of California San Francisco, San Francisco, California
| | - Mark Schumacher
- Department of Anesthesia and Perioperative Care, Division of Pain Medicine, University of California, San Francisco, San Francisco, California
| | - Brett R Stacey
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Thomas Van De Ven
- Duke University Department of Anesthesiology, Division of Pain Medicine, Durham, North Carolina
| |
Collapse
|
104
|
Luo X, Gu Y, Tao X, Serhan CN, Ji RR. Resolvin D5 Inhibits Neuropathic and Inflammatory Pain in Male But Not Female Mice: Distinct Actions of D-Series Resolvins in Chemotherapy-Induced Peripheral Neuropathy. Front Pharmacol 2019; 10:745. [PMID: 31333464 PMCID: PMC6624779 DOI: 10.3389/fphar.2019.00745] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Earlier studies have demonstrated that essential fatty acid-derived specialized pro-resolving mediators (SPMs) promote the resolution of inflammation and pain. However, the potential analgesic actions of SPMs in chemotherapy-induced peripheral neuropathy (CIPN) are not known. Recent results also showed sex dimorphism in immune cell signaling in neuropathic pain. Here, we evaluated the analgesic actions of D-series resolvins (RvD1, RvD2, RvD3, RvD4, and RvD5) on a CIPN in male and female mice. Paclitaxel (PTX, 2 mg/kg), given on days 0, 2, 4, and 6, produced robust mechanical allodynia in both sexes at 2 weeks. Intrathecal injection of RvD1 and RvD2 (100 ng, i.t.) at 2 weeks reversed PTX-induced mechanical allodynia in both sexes, whereas RvD3 and RvD4 (100 ng, i.t.) had no apparent effects on either sex. Interestingly, RvD5 (100 ng, i.t.) only reduced mechanical allodynia in male mice but not in female mice. Notably, PTX-induced mechanical allodynia was fully developed in Trpv1 or Trpa1 knockout mice, showing no sex differences. Also, intrathecal RvD5 reduced mechanical allodynia in male mice lacking Trpv1 or Trpa1, whereas female mice with Trpv1 or Trpa1 deficiency had no response to RvD5. Finally, RvD5-induced male-specific analgesia was also confirmed in an inflammatory pain condition. Formalin-induced second phase pain (licking and flinching) was reduced by intrathecal RvD5 in male but not female mice. These findings identified RvD5 as the first SPM that shows sex dimorphism in pain regulation. Moreover, these results suggest that specific resolvins may be used to treat CIPN, a rising health concern in cancer survivors.
Collapse
Affiliation(s)
- Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Yun Gu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Xueshu Tao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Charles Nicholas Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States.,Department of Neurobiology, Duke University Medical Center, Durham, NC, United States.,Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
105
|
McKay TB, Seyed-Razavi Y, Ghezzi CE, Dieckmann G, Nieland TJF, Cairns DM, Pollard RE, Hamrah P, Kaplan DL. Corneal pain and experimental model development. Prog Retin Eye Res 2019; 71:88-113. [PMID: 30453079 PMCID: PMC6690397 DOI: 10.1016/j.preteyeres.2018.11.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 11/03/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022]
Abstract
The cornea is a valuable tissue for studying peripheral sensory nerve structure and regeneration due to its avascularity, transparency, and dense innervation. Somatosensory innervation of the cornea serves to identify changes in environmental stimuli at the ocular surface, thereby promoting barrier function to protect the eye against injury or infection. Due to regulatory demands to screen ocular safety of potential chemical exposure, a need remains to develop functional human tissue models to predict ocular damage and pain using in vitro-based systems to increase throughput and minimize animal use. In this review, we summarize the anatomical and functional roles of corneal innervation in propagation of sensory input, corneal neuropathies associated with pain, and the status of current in vivo and in vitro models. Emphasis is placed on tissue engineering approaches to study the human corneal pain response in vitro with integration of proper cell types, controlled microenvironment, and high-throughput readouts to predict pain induction. Further developments in this field will aid in defining molecular signatures to distinguish acute and chronic pain triggers based on the immune response and epithelial, stromal, and neuronal interactions that occur at the ocular surface that lead to functional outcomes in the brain depending on severity and persistence of the stimulus.
Collapse
Affiliation(s)
- Tina B McKay
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Yashar Seyed-Razavi
- Center for Translational Ocular Immunology and Cornea Service, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Chiara E Ghezzi
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Gabriela Dieckmann
- Center for Translational Ocular Immunology and Cornea Service, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Thomas J F Nieland
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Rachel E Pollard
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology and Cornea Service, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA.
| |
Collapse
|
106
|
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) and female sexual dysfunction (FSD) are common conditions that substantially reduce women's health. In particular, women with IC/BPS show vulvodynia, a kind of FDS that originates from consistent pain around the vulvar area. There have been many studies attempting to find the underlying mechanisms that induce the chronic pain associated with IC/BPS and vulvodynia and explain why these two conditions often coexist. Proposed theories suggest that pain hypersensitivity is being mediated by peripheral and central sensitization. However, there are still many unknown factors, such as etiologies, that can evoke pain hypersensitivity and may be linking the casual relationship between IC/BPS and vulvodynia. At present, knowledge regarding IC/BPS and vulvodynia are insufficient when considering their clinical importance. Therefore, efforts are necessary to elucidate the issues surrounding IC/BPS and vulvodynia.
Collapse
Affiliation(s)
- Su Jin Kim
- Department of Urology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea.,Current address: Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Jayoung Kim
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA. .,Department of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| | - Hana Yoon
- Department of Urology, Medical Research Center, Ewha Womans University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
107
|
Nociceptor Signalling through ion Channel Regulation via GPCRs. Int J Mol Sci 2019; 20:ijms20102488. [PMID: 31137507 PMCID: PMC6566991 DOI: 10.3390/ijms20102488] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022] Open
Abstract
The prime task of nociceptors is the transformation of noxious stimuli into action potentials that are propagated along the neurites of nociceptive neurons from the periphery to the spinal cord. This function of nociceptors relies on the coordinated operation of a variety of ion channels. In this review, we summarize how members of nine different families of ion channels expressed in sensory neurons contribute to nociception. Furthermore, data on 35 different types of G protein coupled receptors are presented, activation of which controls the gating of the aforementioned ion channels. These receptors are not only targeted by more than 20 separate endogenous modulators, but can also be affected by pharmacotherapeutic agents. Thereby, this review provides information on how ion channel modulation via G protein coupled receptors in nociceptors can be exploited to provide improved analgesic therapy.
Collapse
|
108
|
He Y, Wang ZJ. Spinal and afferent PKC signaling mechanisms that mediate chronic pain in sickle cell disease. Neurosci Lett 2019; 706:56-60. [PMID: 31051220 DOI: 10.1016/j.neulet.2019.04.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
Pain is the most characteristic feature of sickle cell disease (SCD). Patients with SCD live with unpredictable, recurrent episodes of acute painful crisis, as well as chronic unremitting pain throughout their lifetime. While most of the research and medical efforts have focused on treating vaso-occlusion crisis and acute pain, chronic pain remains a significant challenge faced by patients and physicians. Emerging evidence from human and animal studies has suggested the presence of a neuropathic component in SCD pain. New knowledge on the neurobiology of chronic pain in SCD has significant implications in unraveling the underlying mechanisms. This review focuses on the recent advances on the role of protein kinase C or PKC in promoting and maintaining chronic pain conditions. With a highlight of a specific PKC isoform, PKCδ, we aim to propose PKC as an essential regulator of chronic pain in SCD, which may ultimately lead to innovative therapeutic strategies for treating this devastating life-long problem in patients with SCD.
Collapse
Affiliation(s)
- Ying He
- Department of Biopharmaceutical Sciences and Center for Biomolecular Sciences, University of Illinois, Chicago, IL 60612, United States.
| | - Zaijie Jim Wang
- Department of Biopharmaceutical Sciences and Center for Biomolecular Sciences, University of Illinois, Chicago, IL 60612, United States.
| |
Collapse
|
109
|
Yin Y, Guo R, Shao Y, Ge M, Miao C, Cao L, Yang Y, Hu L. Pretreatment with resveratrol ameliorate trigeminal neuralgia by suppressing matrix metalloproteinase-9/2 in trigeminal ganglion. Int Immunopharmacol 2019; 72:339-347. [PMID: 31009895 DOI: 10.1016/j.intimp.2019.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/29/2019] [Accepted: 04/05/2019] [Indexed: 01/05/2023]
Abstract
Trigeminal neuralgia (TN) is a common type of neuropathic pain whereas the underlying pathogenesis has not been completely elucidated. Recent study suggests that the development of neuroinflammation is responsible for generating and sustaining neuropathic pain. The purpose of our study was to investigate the protective effect of intervening the inflammation in early stages of pain and explore its potential mechanism. MMP-9 and MMP-2 are vital proinflammatory participants and accumulating evidence indicates that they are involved in the early development of neuropathic pain. In this study, we found that MMP-9/2 showed different temporal up regulation in trigeminal ganglion (TG) significantly after chronic constriction injury (CCI) surgery. However, the activation of MMP-9/2 were suppressed by the pretreatment with resveratrol, which delayed and attenuated CCI-induced mechanical allodynia simultaneously. Besides, the expression of proinflammatory cytokines like IL-1β and TNF-α as well as the excessive neuronal activity induced by CCI were suppressed by resveratrol. Moreover, we believed that the inhibition of MMP-9/2 activation and pain sensitization may be related to the TLR-4/NF-κB signaling pathway, which might be negatively regulated by the induction of SOCS3. In conclusion, pretreatment with resveratrol could be an effective approach to alleviate trigeminal neuralgia in early stages via a powerful inhibition on the activation of MMP-9/2 in TG.
Collapse
Affiliation(s)
- Yuling Yin
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Rong Guo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yu Shao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Mixue Ge
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Chen Miao
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Ling Cao
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yanjing Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China; Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.
| | - Liang Hu
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.
| |
Collapse
|
110
|
Malange KF, Dos Santos GG, Kato NN, Toffoli-Kadri MC, Carollo CA, Silva DB, Portugal LC, Alves FM, Rita PHS, Parada CA, Rondon ES. Tabebuia aurea decreases hyperalgesia and neuronal injury induced by snake venom. JOURNAL OF ETHNOPHARMACOLOGY 2019; 233:131-140. [PMID: 30590196 DOI: 10.1016/j.jep.2018.12.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tabebuia aurea (Silva Manso) Benth. & Hook. f. ex S. Moore is used as anti-inflammatory, analgesic and antiophidic in traditional medicine, though its pharmacological proprieties are still underexplored. In the bothropic envenoming, pain is a key symptom drove by an intense local inflammatory and neurotoxic event. The antivenom serum therapy is still the main treatment despite its poor local effects against pain and tissue injury. Furthermore, it is limited to ambulatorial niches, giving space for the search of new and more inclusive pharmacological approaches. AIM OF THE STUDY evaluation of Tabebuia aurea hydroethanolic extract (HEETa) in hyperalgesia and neuronal injury induced by Bothrops mattogrossensis venom (VBm). MATERIALS AND METHODS Stem barks from Tabebuia aurea were extracted with ethanol and water (7:3, v/v) to yield the extract HEETa. Then, HEETa was analyzed by LC-DAD-MS and its constituents were identified. Snake venoms were extracted from adult specimens of Bothrops mattogrossensis, lyophilized and kept at -20 °C until use. Male Swiss mice, weighting 20-25 g, were used to hyperalgesia (electronic von Frey), motor impairment (Rotarod test) and tissue injury evaluation (histopatology and ATF-3 immunohistochemistry). Therefore, three experimental groups were formed: VBm (1 pg, 1 ng, 0.3 μg, 1 μg, 3 and 6 μg/paw), HEETa orally (180, 540, 720, 810 or 1080 mg/kg; 10 mL/kg, 30 min prior VBm inoculation) and VBm neutralized (VBm: HEETa, 1:100 parts, respectively). In all set of experiments a control (saline group) was used. First, we made a dose-time-response course curve of VBm's induced hyperalgesia. Next, VBm maximum hyperalgesic dose was employed to perform HEETa orally dose-time-response course curve and analyses of VBm neutralized. Paw tissues for histopathology and DRGs were collected from animals inoculated with VBm maximum dose and treated with HEETa antihyperalgesic effective dose or neutralized VBm. Paws were extract two or 72 h after VBm inoculation and DRGs, in the maximum expected time expression of ATF-3 (72 h). RESULTS From HEETa extract, glycosylated iridoids were identified, such as catalpol, minecoside, verminoside and specioside. VBm induced a time and dose dependent hyperalgesia with its highest effect seen with 3 µg/paw, 2 h after venom inoculation. HEETa effective dose (720 mg/kg) decreased significantly VBm induced hyperalgesia (3 µg/paw) with no motor impairment and signs of acute toxicity. HEETa antihyperalgesic action starts 1.5 h after VBm inoculation and lasted up until 2 h after VBm. Hyperalgesia wasn't reduced by VBm: HEETa neutralization. Histopathology revealed a large hemorragic field 2 h after VBm inoculation and an intense inflammatory infiltrate of polymorphonuclear cells at 72 h. Both HEETa orally and VBm: HEETa groups had a reduced inflammation at 72 h after VBm. Also, the venom significantly induced ATF-3 expression (35.37 ± 3.25%) compared with saline group (4.18 ± 0.68%) which was reduced in HEETa orally (25.87 ± 2.57%) and VBm: HEETa (19.84 ± 2.15%) groups. CONCLUSION HEETa reduced the hyperalgesia and neuronal injury induced by VBm. These effects could be related to iridoid glycosides detected in HEETa and their intrinsic reported mechanism.
Collapse
Affiliation(s)
- Kauê Franco Malange
- Faculty of Pharmaceutical Sciences, Food, and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Gilson Gonçalves Dos Santos
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Natália Naomi Kato
- Faculty of Pharmaceutical Sciences, Food, and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Mônica Cristina Toffoli-Kadri
- Faculty of Pharmaceutical Sciences, Food, and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Carlos Alexandre Carollo
- Faculty of Pharmaceutical Sciences, Food, and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Denise Brentan Silva
- Faculty of Pharmaceutical Sciences, Food, and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Luciane Candeloro Portugal
- Institute of Biosciences, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Flávio Macedo Alves
- Institute of Biosciences, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | | | - Carlos Amílcar Parada
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Eric Schmidt Rondon
- Faculty of Veterinary Medicine and Animal Sciences (FAMEZ), Federal University of Mato Grosso do Sul, Department of Veterinary Medicine, Campo Grande, Mato Grosso do Sul, Brazil.
| |
Collapse
|
111
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
112
|
Goldstein RH, Barkai O, Íñigo-Portugués A, Katz B, Lev S, Binshtok AM. Location and Plasticity of the Sodium Spike Initiation Zone in Nociceptive Terminals In Vivo. Neuron 2019; 102:801-812.e5. [PMID: 30926280 DOI: 10.1016/j.neuron.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 01/15/2019] [Accepted: 03/01/2019] [Indexed: 11/18/2022]
Abstract
Nociceptive terminals possess the elements for detecting, transmitting, and modulating noxious signals, thus being pivotal for pain sensation. Despite this, a functional description of the transduction process by the terminals, in physiological conditions, has not been fully achieved. Here, we studied how nociceptive terminals in vivo convert noxious stimuli into propagating signals. By monitoring noxious-stimulus-induced Ca2+ dynamics from mouse corneal terminals, we found that initiation of Na+ channel (Nav)-dependent propagating signals takes place away from the terminal and that the starting point for Nav-mediated propagation depends on Nav functional availability. Acute treatment with the proinflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β) resulted in a shift of the location of Nav involvement toward the terminal, thus increasing nociceptive excitability. Moreover, a shift of Nav involvement toward the terminal occurs in corneal hyperalgesia resulting from acute photokeratitis. This dynamic change in the location of Nav-mediated propagation initiation could underlie pathological pain hypersensitivity.
Collapse
Affiliation(s)
- Robert H Goldstein
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, 9112001 Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, 9112001 Jerusalem, Israel
| | - Omer Barkai
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, 9112001 Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, 9112001 Jerusalem, Israel
| | - Almudena Íñigo-Portugués
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Ben Katz
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, 9112001 Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, 9112001 Jerusalem, Israel
| | - Shaya Lev
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, 9112001 Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, 9112001 Jerusalem, Israel
| | - Alexander M Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University, 9112001 Jerusalem, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, 9112001 Jerusalem, Israel.
| |
Collapse
|
113
|
Tracey I, Woolf CJ, Andrews NA. Composite Pain Biomarker Signatures for Objective Assessment and Effective Treatment. Neuron 2019; 101:783-800. [PMID: 30844399 PMCID: PMC6800055 DOI: 10.1016/j.neuron.2019.02.019] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/05/2019] [Accepted: 02/13/2019] [Indexed: 02/09/2023]
Abstract
Pain is a subjective sensory experience that can, mostly, be reported but cannot be directly measured or quantified. Nevertheless, a suite of biomarkers related to mechanisms, neural activity, and susceptibility offer the possibility-especially when used in combination-to produce objective pain-related indicators with the specificity and sensitivity required for diagnosis and for evaluation of risk of developing pain and of analgesic efficacy. Such composite biomarkers will also provide improved understanding of pain pathophysiology.
Collapse
Affiliation(s)
- Irene Tracey
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| | - Clifford J Woolf
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, 02115 MA, USA.
| | - Nick A Andrews
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, 02115 MA, USA
| |
Collapse
|
114
|
Sengupta S, Mehta G. Natural products as modulators of the cyclic-AMP pathway: evaluation and synthesis of lead compounds. Org Biomol Chem 2019; 16:6372-6390. [PMID: 30140804 DOI: 10.1039/c8ob01388h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is now well recognized that the normal cellular response in mammalian cells is critically regulated by the cyclic-AMP (cAMP) pathway through the appropriate balance of adenylyl cyclase (AC) and phosphodiesterase-4 (PDE4) activities. Dysfunctions in the cAMP pathway have major implications in various diseases like CNS disorders, inflammation and cardiac syndromes and, hence, the modulation of cAMP signalling through appropriate intervention of AC/PDE4 activities has emerged as a promising new drug discovery strategy of current interest. In this context, synthetic small molecules have had limited success so far and therefore parallel efforts on natural product leads have been actively pursued. The early promise of using the diterpene forskolin and its semi-synthetic analogs as AC activators has given way to new leads in the last decade from novel natural products like the marine sesterterpenoids alotaketals and ansellones and the 9,9'-diarylfluorenone cored selaginpulvilins, etc. and their synthesis has drawn much attention. This review captures these contemporary developments, particularly total synthesis campaigns and structure-guided analog design in the context of AC and PDE-4 modulating attributes and the scope for future possibilities.
Collapse
Affiliation(s)
- Saumitra Sengupta
- School of Chemistry, University of Hyderabad, Gachibowli, Hyderabad - 5000 046, Telengana, India.
| | | |
Collapse
|
115
|
Brönnimann BVE, Hou MY, Zembic A, Parkinson CX, Meier ML, Ettlin DA. Dentin hypersensitivity monitored by cold air quantitative sensory testing. J Oral Rehabil 2019; 46:549-555. [PMID: 30802997 DOI: 10.1111/joor.12781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Quantification of dentin hypersensitivity (DH) is challenging and requires standardised, graded stimulation by natural-like stimuli. OBJECTIVE The present study aimed at identifying DH subjects and longitudinally monitoring their pain thresholds by cold air quantitative sensory testing (QST). METHODS Subject recruitment started with an online DH questionnaire. Respondents were screened by dental air stimulation. Sensitising and habituating subjects were excluded. A recently developed stimulation device was employed for cold air QST. Single tooth DH was verified by applying an equi-intense stimulus to a control tooth. Descriptive statistics were applied for subject characteristics. Mean values were calculated for the stimulation parameters temperature and air flow. Reliability of temperatures for detecting pain and for evoking moderate pain over multiple time points within a 3-week period was analysed by two-way random single- and average-measure intra-class correlation coefficients. RESULTS A total of 353 persons completed the online DH questionnaire of which 117 were screened. Forty-four passed the screening, yet 15 were excluded for various reasons. Twenty-nine subjects were monitored by QST across 3 weeks. Results revealed a high intra-individual stability of the temperature inducing moderate to strong pain intensity (MPI) (single-measure ICC of TMPI 0.83, P < 0.001). Mean TMPI was -13.69°C, yet it highly varied among the 29 subjects (SD ± 10.04°C). CONCLUSIONS Using a novel approach, namely dental QST based on cold air stimuli, we present evidence for temporally stable DH perceptions over a 3-week period. The method fulfils international guideline requirements and is recommendable for obtaining valid results when testing various interventions for DH management.
Collapse
Affiliation(s)
- Ben V E Brönnimann
- Ambulatory Center for Psychiatry and Psychotherapy, Psychiatric Services of District Aargau, Aargau, Switzerland
| | - Mei-Yin Hou
- Center of Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Anja Zembic
- Center of Dental Medicine, University of Zurich, Zurich, Switzerland.,Zahnmedizin Zurich Nord, Zurich, Switzerland
| | | | - Michael L Meier
- Center of Dental Medicine, University of Zurich, Zurich, Switzerland.,Balgrist University Hospital, Zurich, Switzerland
| | - Dominik A Ettlin
- Center of Dental Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
116
|
Xiang HC, Lin LX, Hu XF, Zhu H, Li HP, Zhang RY, Hu L, Liu WT, Zhao YL, Shu Y, Pan HL, Li M. AMPK activation attenuates inflammatory pain through inhibiting NF-κB activation and IL-1β expression. J Neuroinflammation 2019; 16:34. [PMID: 30755236 PMCID: PMC6373126 DOI: 10.1186/s12974-019-1411-x] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 01/22/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Chronic pain is a major clinical problem with limited treatment options. Previous studies have demonstrated that activation of adenosine monophosphate-activated protein kinase (AMPK) can attenuate neuropathic pain. Inflammation/immune response at the site of complete Freund's adjuvant (CFA) injection is known to be a critical trigger of the pathological changes that produce inflammatory pain. However, whether activation of AMPK produces an analgesic effect through inhibiting the proinflammatory cytokines, including interleukin-1β (IL-1β), in inflammatory pain remains unknown. METHODS Inflammatory pain was induced in mice injected with CFA. The effects of AICAR (5-aminoimidazole-4-carboxyamide ribonucleoside, an AMPK activator), Compound C (an AMPK inhibitor), and IL-1ra (an IL-1 receptor antagonist) were tested at day 4 after CFA injection. Inflammatory pain was assessed with von Frey filaments and hot plate. Immunoblotting, hematoxylin and eosin (H&E) staining, and immunofluorescence were used to assess inflammation-induced biochemical changes. RESULTS The AMPK activator AICAR produced an analgesic effect and inhibited the level of proinflammatory cytokine IL-1β in the inflamed skin in mice. Moreover, activation of AMPK suppressed CFA-induced NF-κB p65 translocation from the cytosol to the nucleus in activated macrophages (CD68+ and CX3CR1+) of inflamed skin tissues. Subcutaneous injection of IL-1ra attenuated CFA-induced inflammatory pain. The AMPK inhibitor Compound C and AMPKα shRNA reversed the analgesic effect of AICAR and the effects of AICAR on IL-1β and NF-κB activation in inflamed skin tissues. CONCLUSIONS Our study provides new information that AMPK activation produces the analgesic effect by inhibiting NF-κB activation and reducing the expression of IL-1β in inflammatory pain.
Collapse
Affiliation(s)
- Hong-Chun Xiang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China
| | - Li-Xue Lin
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China
| | - Xue-Fei Hu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China
| | - He Zhu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China
| | - Hong-Ping Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China
| | - Ru-Yue Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China
| | - Liang Hu
- Department of Pharmacology, School of Basic Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - Wen-Tao Liu
- Department of Pharmacology, School of Basic Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - Yi-Lin Zhao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Shu
- Department of Central Laboratory, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Hui-Lin Pan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Man Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
117
|
Komatsu R, Carvalho B, Flood P. Prediction of outliers in pain, analgesia requirement, and recovery of function after childbirth: a prospective observational cohort study. Br J Anaesth 2019; 121:417-426. [PMID: 30032880 DOI: 10.1016/j.bja.2018.04.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/09/2018] [Accepted: 05/02/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Prediction models to identify parturients who experience protracted pain, prolonged opioid use, and delayed self-assessed functional recovery are currently inadequate. METHODS For this study, 213 nulliparous women who planned vaginal delivery were enrolled and assessed daily until they completed three outcomes: (1) pain resolution; (2) opioid cessation; and (3) self-assessed functional recovery to predelivery level. The primary composite endpoint, 'pain and opioid-free functional recovery' was the time required to reach all three endpoints. The subjects were divided into two categories (the worst (longest time) 20% and remaining 80%) for reaching the primary composite endpoint, and each individual component. Prediction models for prolonged recovery were constructed using multivariate logistic regression with demographic, obstetric, psychological, and health-related quality of life characteristics as candidate predictors. RESULTS Labour induction (vs spontaneous labour onset) predicted the worst 20% for the primary composite endpoint in the final multivariate model. Labour induction and higher postpartum day 1 numerical rating score for pain were predictors for being in the worst 20% for both functional recovery and pain burden. Labour type, delivery type, Patient-Reported Outcomes Measurement Information System (PROMIS) anxiety score, RAND 36 Item Health Survey 1.0 (SF-36) physical health composite score, and postpartum breastfeeding success were predictive of delayed opioid cessation. CONCLUSIONS Labour induction and elevated numerical rating score for pain are predictive of poor recovery after childbirth. Further research is necessary to determine whether modification would benefit mothers at risk for poor recovery.
Collapse
Affiliation(s)
- R Komatsu
- Department of Anaesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| | - B Carvalho
- Department of Anaesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - P Flood
- Department of Anaesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
118
|
Spinal Serotonin 1A Receptor Contributes to the Analgesia of Acupoint Catgut Embedding by Inhibiting Phosphorylation of the N-Methyl-d-Aspartate Receptor GluN1 Subunit in Complete Freund's Adjuvant-Induced Inflammatory Pain in Rats. THE JOURNAL OF PAIN 2019; 20:16.e1-16.e16. [DOI: 10.1016/j.jpain.2018.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/21/2018] [Accepted: 07/24/2018] [Indexed: 02/08/2023]
|
119
|
Abstract
Increasing numbers of those living with and beyond cancer presents a clinical challenge for pain specialists. A large proportion of these patients experience pain secondary to their disease or its treatment, impeding rehabilitation and significantly impacting upon their quality of life. The successful management of this pain presents a considerable challenge. This review aims to outline current concepts and treatment options, while considering nuances within pain assessment and the use of large-scale data to help guide further advances.
Collapse
Affiliation(s)
- David Magee
- Department of Pain Medicine, The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Sabina Bachtold
- Department of Pain Medicine, The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Matthew Brown
- Department of Pain Medicine, The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK.,Targeted Approaches to Cancer Pain Group, The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK
| | - Paul Farquhar-Smith
- Department of Pain Medicine, The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
120
|
Pan F, Jones G. Clinical Perspective on Pain and Pain Phenotypes in Osteoarthritis. Curr Rheumatol Rep 2018; 20:79. [DOI: 10.1007/s11926-018-0796-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
121
|
Farajzadeh A, Bathaie SZ, Arabkheradmand J, Ghodsi SM, Faghihzadeh S. Different Pain States of Trigeminal Neuralgia Make Significant Changes in the Plasma Proteome and Some Biochemical Parameters: a Preliminary Cohort Study. J Mol Neurosci 2018; 66:524-534. [DOI: 10.1007/s12031-018-1183-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 09/24/2018] [Indexed: 01/03/2023]
|
122
|
Abstract
Abstract Primary sensory neurons are responsible for transmitting sensory information from the peripheral to the central nervous system. Their responses to incoming stimulation become greatly enhanced and prolonged following inflammation, giving rise to exaggerated nociceptive responses and chronic pain. The inflammatory mediator, prostaglandin E2 (PGE2), released from the inflamed tissue surrounding the terminals of sensory neurons contributes to the abnormal pain responses. PGE2 acts on G protein-coupled EP receptors to activate adenylyl cyclase, which catalyzes the conversion of adenosine triphosphate to cyclic adenosine 3′,5′-monophosphate (cAMP). Under normal conditions, cAMP activates primarily protein kinase A. After inflammation, cAMP also activates the exchange proteins activated by cAMP (Epacs) to produce exaggerated PGE2-mediated hyperalgesia. The role of cAMP-Epac signaling in the generation of hypersensitivity is the topic of this review.
Collapse
Affiliation(s)
| | - Yanping Gu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch Galveston, TX 77555-1069, USA
| |
Collapse
|
123
|
Strand J, Stinson C, Bellinger LL, Peng Y, Kramer PR. G i protein functions in thalamic neurons to decrease orofacial nociceptive response. Brain Res 2018; 1694:63-72. [PMID: 29763576 PMCID: PMC6026072 DOI: 10.1016/j.brainres.2018.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/27/2018] [Accepted: 05/12/2018] [Indexed: 01/01/2023]
Abstract
Orofacial pain includes neuronal pathways that project from the trigeminal nucleus to and through the thalamus. What role the ventroposterior thalamic complex (VP) has on orofacial pain transmission is not understood. To begin to address this question an inhibitory G protein (Gi) designer receptor exclusively activated by a designer drug (DREADD) was transfected in cells of the VP using adeno-associated virus isotype 8. Virus infected cells were identified by a fluorescent tag and immunostaining. Cells were silenced after injecting the designer drug clozapine-n-oxide, which binds the designer receptor activating Gi. Facial rubbing and local field potentials (LFP) in the VP were then recorded in awake, free moving Sprague Dawley rats after formalin injection of the masseter muscle to induce nociception. Formalin injection significantly increased LFP and the nociceptive behavioral response. Activation of DREADD Gi with clozapine-n-oxide significantly reduced LFP in the VP and reduced the orofacial nociceptive response. Because DREADD silencing can result from Gi-coupled inwardly-rectifying potassium channels (GIRK), the GIRK channel blocker tertiapin-Q was injected. Injection of GIRK blocker resulted in an increase in the nociceptive response and increased LFP activity. Immunostaining of the VP for glutamate vesicular transporter (VGLUT2) and gamma-aminobutyric acid vesicular transporter (VGAT) indicated a majority of the virally transfected cells were excitatory (VGLUT2 positive) and a minority were inhibitory (VGAT positive). We conclude first, that inhibition of the excitatory neurons within the VP reduced electrical activity and the orofacial nociceptive response and that the effect on excitatory neurons overwhelmed any change resulting from inhibitor neurons. Second, inhibition of LFP and nociception was due, in part, to GIRK activation.
Collapse
Affiliation(s)
- Jennifer Strand
- Department of Psychology, University of Texas at Arlington, Arlington, TX 76019, United States
| | - Crystal Stinson
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States
| | - Larry L Bellinger
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States
| | - Yuan Peng
- Department of Psychology, University of Texas at Arlington, Arlington, TX 76019, United States
| | - Phillip R Kramer
- Texas A&M University College of Dentistry, Dallas, TX 75246, United States.
| |
Collapse
|
124
|
Ji RR, Nackley A, Huh Y, Terrando N, Maixner W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 2018; 129:343-366. [PMID: 29462012 PMCID: PMC6051899 DOI: 10.1097/aln.0000000000002130] [Citation(s) in RCA: 769] [Impact Index Per Article: 128.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic pain is maintained in part by central sensitization, a phenomenon of synaptic plasticity, and increased neuronal responsiveness in central pain pathways after painful insults. Accumulating evidence suggests that central sensitization is also driven by neuroinflammation in the peripheral and central nervous system. A characteristic feature of neuroinflammation is the activation of glial cells, such as microglia and astrocytes, in the spinal cord and brain, leading to the release of proinflammatory cytokines and chemokines. Recent studies suggest that central cytokines and chemokines are powerful neuromodulators and play a sufficient role in inducing hyperalgesia and allodynia after central nervous system administration. Sustained increase of cytokines and chemokines in the central nervous system also promotes chronic widespread pain that affects multiple body sites. Thus, neuroinflammation drives widespread chronic pain via central sensitization. We also discuss sex-dependent glial/immune signaling in chronic pain and new therapeutic approaches that control neuroinflammation for the resolution of chronic pain.
Collapse
Affiliation(s)
- Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710
| | - Andrea Nackley
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
125
|
Giblin JP, Etayo I, Castellanos A, Andres-Bilbe A, Gasull X. Anionic Phospholipids Bind to and Modulate the Activity of Human TRESK Background K + Channel. Mol Neurobiol 2018; 56:2524-2541. [PMID: 30039335 DOI: 10.1007/s12035-018-1244-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/15/2018] [Indexed: 01/08/2023]
Abstract
The background K+ channel TRESK regulates sensory neuron excitability, and changes in its function/expression contribute to neuronal hyperexcitability after injury/inflammation, making it an attractive therapeutic target for pain-related disorders. Factors that change lipid bilayer composition/properties (including volatile anesthetics, chloroform, chlorpromazine, shear stress, and cell swelling/shrinkage) modify TRESK current, but despite the importance of anionic phospholipids (e.g., PIP2) in the regulation of many ion channels, it remains unknown if membrane lipids affect TRESK function. We describe that both human and rat TRESK contain potential anionic phospholipid binding sites (apbs) in the large cytoplasmic loop, but only the human channel is able to bind to multilamellar vesicles (MLVs), enriched with anionic phospholipids, suggesting an electrostatically mediated interaction. We mapped the apbs to a short stretch of 14 amino acids in the loop, located at the membrane-cytosol interface. Disruption of electrostatic lipid-TRESK interactions inhibited hTRESK currents, while subsequent application of Folch Fraction MLVs or a PIP2 analog activated hTRESK, an effect that was absent in the rat ortholog. Strikingly, channel activation by anionic phospholipids was conferred to rTRESK by replacing the equivalent rat sequence with the human apbs. Finally, in the presence of a calcineurin inhibitor, stimulation of a Gq/11-linked GPCR reduced hTRESK current, revealing a likely inhibitory effect of membrane lipid hydrolysis on hTRESK activity. This novel regulation of hTRESK by anionic phospholipids is a characteristic of the human channel that is not present in rodent orthologs. This must be considered when extrapolating results from animal models and may open the door to the development of novel channel modulators as analgesics.
Collapse
Affiliation(s)
- Jonathan P Giblin
- Neurophysiology Lab, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Iñigo Etayo
- Neurophysiology Lab, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Aida Castellanos
- Neurophysiology Lab, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Alba Andres-Bilbe
- Neurophysiology Lab, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Xavier Gasull
- Neurophysiology Lab, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.
| |
Collapse
|
126
|
Long-Term Outcomes in the Management of Central Neuropathic Pain Syndromes: A Prospective Observational Cohort Study. Can J Neurol Sci 2018; 45:545-552. [DOI: 10.1017/cjn.2018.55] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractBackground:Central neuropathic pain syndromes are a result of central nervous system injury, most commonly related to stroke, traumatic spinal cord injury, or multiple sclerosis. These syndromes are distinctly less common than peripheral neuropathic pain, and less is known regarding the underlying pathophysiology, appropriate pharmacotherapy, and long-term outcomes. The objective of this study was to determine the long-term clinical effectiveness of the management of central neuropathic pain relative to peripheral neuropathic pain at tertiary pain centers.Methods:Patients diagnosed with central (n=79) and peripheral (n=710) neuropathic pain were identified for analysis from a prospective observational cohort study of patients with chronic neuropathic pain recruited from seven Canadian tertiary pain centers. Data regarding patient characteristics, analgesic use, and patient-reported outcomes were collected at baseline and 12-month follow-up. The primary outcome measure was the composite of a reduction in average pain intensity and pain interference. Secondary outcome measures included assessments of function, mood, quality of life, catastrophizing, and patient satisfaction.Results:At 12-month follow-up, 13.5% (95% confidence interval [CI], 5.6-25.8) of patients with central neuropathic pain and complete data sets (n=52) achieved a ≥30% reduction in pain, whereas 38.5% (95% CI, 25.3-53.0) achieved a reduction of at least 1 point on the Pain Interference Scale. The proportion of patients with central neuropathic pain achieving both these measures, and thus the primary outcome, was 9.6% (95% CI, 3.2-21.0). Patients with peripheral neuropathic pain and complete data sets (n=463) were more likely to achieve this primary outcome at 12 months (25.3% of patients; 95% CI, 21.4-29.5) (p=0.012).Conclusion:Patients with central neuropathic pain syndromes managed in tertiary care centers were less likely to achieve a meaningful improvement in pain and function compared with patients with peripheral neuropathic pain at 12-month follow-up.
Collapse
|
127
|
Loeser J, Blunk JA, Ruschulte H, Knitsch J, Karst M, Hucho T. The beta-adrenergic receptor agonist, terbutaline, reduces UVB-induced mechanical sensitization in humans. Eur J Pain 2018; 23:72-80. [PMID: 29984439 DOI: 10.1002/ejp.1286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2018] [Indexed: 11/10/2022]
Abstract
OBJECTIVES Previously, we found in cultures of primary neurons and in animals that sensitized primary neurons can be desensitized by treatment with e.g. beta-adrenergic receptor agonists. We now tested whether also in human sensitization such as UVB-radiation induced sunburn-like hyperalgesia can be reduced by intradermal injection of the beta-adrenergic receptor agonist terbutaline. METHODS In our prospective randomized study, 17 participants received an individual UVB dose to cause a defined local sunburn-like erythema at four locations, two on each forearm. Twenty-four hours later, the sensitized four areas were injected intradermally with terbutaline pH 4.3, terbutaline pH 7.0, saline pH 4.3 or saline pH 7.0, respectively. Pain thresholds were examined before and after induction of UVB-sensitization, and 15, 30 and 60 min after injection of the respective solution. Mechanical pain thresholds of the skin and of deeper tissues were determined by pinprick and pressure algometer measurements, respectively. RESULTS UVB-irradiation decreased mechanical pain thresholds for pinprick and pressure algometer measurements demonstrating a successful sunburn-like sensitization. Intradermal injection of terbutaline pH 7.0 into the sensitized skin reduced the sensitization for all measured timepoints as determined by pinprick measurements. Pinprick measurements of sensitization were not reduced by injection of terbutaline pH 4.3, saline solution pH 7.0 or saline solution pH 4.3. Also, sensitization of deeper tissue nociceptors were not altered by any of the injections as measured with the pressure algometer. CONCLUSIONS Similar to our cellular observations, also in humans beta-adrenergic agonists such as terbutaline can reduce the sensitization of primary neurons in the skin. SIGNIFICANCE We previously showed in model systems that beta-adrenergic stimulation can not only sensitize but also desensitize nociceptors. Our study shows that also in humans beta-adrenergic agonists desensitize if injected into UVB-sensitized skin. This indicates an analgesic activity of adrenergic agonists in addition to their vasoconstrictory function.
Collapse
Affiliation(s)
- J Loeser
- Department of Anaesthesiology and Intensive Care Medicine, University Clinic of Cologne, Cologne, Germany
| | - J A Blunk
- Department of Pain Therapy, Hospital zum Heiligen Geist GmbH, Kempen, Germany
| | - H Ruschulte
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - J Knitsch
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - M Karst
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - T Hucho
- Department of Anaesthesiology and Intensive Care Medicine, University Clinic of Cologne, Cologne, Germany
| |
Collapse
|
128
|
Aczél T, Kun J, Szőke É, Rauch T, Junttila S, Gyenesei A, Bölcskei K, Helyes Z. Transcriptional Alterations in the Trigeminal Ganglia, Nucleus and Peripheral Blood Mononuclear Cells in a Rat Orofacial Pain Model. Front Mol Neurosci 2018; 11:219. [PMID: 29997476 PMCID: PMC6028693 DOI: 10.3389/fnmol.2018.00219] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022] Open
Abstract
Orofacial pain and headache disorders are among the most debilitating pain conditions. While the pathophysiological basis of these disorders may be diverse, it is generally accepted that a common mechanism behind the arising pain is the sensitization of extra- and intracranial trigeminal primary afferents. In the present study we investigated gene expression changes in the trigeminal ganglia (TRG), trigeminal nucleus caudalis (TNC) and peripheral blood mononuclear cells (PBMC) evoked by Complete Freund's Adjuvant (CFA)-induced orofacial inflammation in rats, as a model of trigeminal sensitization. Microarray analysis revealed 512 differentially expressed genes between the ipsi- and contralateral TRG samples 7 days after CFA injection. Time-dependent expression changes of G-protein coupled receptor 39 (Gpr39), kisspeptin-1 receptor (Kiss1r), kisspeptin (Kiss1), as well as synaptic plasticity-associated Lkaaear1 (Lkr) and Neurod2 mRNA were described on the basis of qPCR results. The greatest alterations were observed on day 3 ipsilaterally, when orofacial mechanical allodynia reached its maximum. This corresponded well with patterns of neuronal (Fosb), microglia (Iba1), and astrocyte (Gfap) activation markers in both TRG and TNC, and interestingly also in PBMCs. This is the first description of up- and downregulated genes both in primary and secondary sensory neurones of the trigeminovascular system that might play important roles in neuroinflammatory activation mechanisms. We are the first to show transcriptomic alterations in the PBMCs that are similar to the neuronal changes. These results open new perspectives and initiate further investigations in the research of trigeminal pain disorders.
Collapse
Affiliation(s)
- Timea Aczél
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - József Kun
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- MTA-PTE Chronic Pain Research Group, Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- MTA-PTE Chronic Pain Research Group, Pécs, Hungary
| | - Tibor Rauch
- Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Sini Junttila
- Bioinformatics and Scientific Computing, Vienna Biocenter Core Facilities, Vienna, Austria
| | - Attila Gyenesei
- Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- Bioinformatics and Scientific Computing, Vienna Biocenter Core Facilities, Vienna, Austria
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre and Centre for Neuroscience, University of Pécs, Pécs, Hungary
- MTA-PTE Chronic Pain Research Group, Pécs, Hungary
| |
Collapse
|
129
|
Faoro MW, Olinto MTA, Paniz VMV, Macagnan J, Henn RL, Garcez A, Pattussi MP. Work-related musculoskeletal pain and its association with common mental disorders among employees of a poultry producing company in Southern Brazil. Rev Bras Med Trab 2018; 16:136-144. [PMID: 32270078 DOI: 10.5327/z1679443520180200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/26/2018] [Indexed: 01/22/2023] Open
Abstract
Background Musculoskeletal pain is one of the most common occupational problems in the industrial society and its prevalence is potentially associated with mental disorders. Objective To estimate the prevalence of work-related musculoskeletal pain and its association with occurrence of common mental disorders among employees of a poultry processing company in Southern Brazil. Methods Cross-sectional study conducted in 2010 with 1,103 employees aged 18 to 52 years old. Musculoskeletal pain was investigated based on a human figure adapted from the Standardized Nordic Questionnaire. We considered reported work-related pain in any part of the body in the past 12 months. Occurrence of common mental disorders was assessed- by the Self-Reporting Questionnaire (SRQ-20). Crude and adjusted prevalence ratios (PR) and corresponding 95% confidence interval (95%CI) were obtained by Poisson regression with robust variance. Results The prevalence of work-related musculoskeletal pain was 40.3% (95%CI 37.4-43.2) for the total sample, 46.8% (95%CI 43.2-50.5) for women and 27.8% (95%CI 23.2-32.3) for men. The prevalence of musculoskeletal pain was twice higher for the participants with common mental disorders compared to those without this condition (PR=2.27; 95%CI 1.99-2.58). This effect remained significant after adjustment for sociodemographic, behavioral, health-related and occupational variables. Conclusion The results of the present study point to the relevance of preventive measures to promote the mental and physical health of workers in order to reduce or minimize the occurrence of pain.
Collapse
Affiliation(s)
- Mariana Wentz Faoro
- Graduate Program in Collective Health, University of the Sinos River Valley (Universidade do Vale do Rio dos Sinos - UNISINOS) - São Leopoldo (RS), Brazil
| | - Maria Teresa Anselmo Olinto
- Graduate Program in Collective Health, University of the Sinos River Valley (Universidade do Vale do Rio dos Sinos - UNISINOS) - São Leopoldo (RS), Brazil
| | - Vera Maria Vieira Paniz
- Graduate Program in Collective Health, University of the Sinos River Valley (Universidade do Vale do Rio dos Sinos - UNISINOS) - São Leopoldo (RS), Brazil
| | - Jamile Macagnan
- Department of Nursing, State University of Santa Catarina (Universidade do Estado de Santa Catarina - UDESC) - Palmitos (SC), Brazil
| | - Ruth Liane Henn
- Graduate Program in Collective Health, University of the Sinos River Valley (Universidade do Vale do Rio dos Sinos - UNISINOS) - São Leopoldo (RS), Brazil
| | - Anderson Garcez
- Graduate Program in Collective Health, University of the Sinos River Valley (Universidade do Vale do Rio dos Sinos - UNISINOS) - São Leopoldo (RS), Brazil
| | - Marcos Pascoal Pattussi
- Graduate Program in Collective Health, University of the Sinos River Valley (Universidade do Vale do Rio dos Sinos - UNISINOS) - São Leopoldo (RS), Brazil
| |
Collapse
|
130
|
Protein kinase A regulates inflammatory pain sensitization by modulating HCN2 channel activity in nociceptive sensory neurons. Pain 2018; 158:2012-2024. [PMID: 28767511 DOI: 10.1097/j.pain.0000000000001005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Several studies implicated cyclic adenosine monophosphate (cAMP) as an important second messenger for regulating nociceptor sensitization, but downstream targets of this signaling pathway which contribute to neuronal plasticity are not well understood. We used a Cre/loxP-based strategy to disable the function of either HCN2 or PKA selectively in a subset of peripheral nociceptive neurons and analyzed the nociceptive responses in both transgenic lines. A near-complete lack of sensitization was observed in both mutant strains when peripheral inflammation was induced by an intradermal injection of 8br-cAMP. The lack of HCN2 as well as the inhibition of PKA eliminated the cAMP-mediated increase of calcium transients in dorsal root ganglion neurons. Facilitation of Ih via cAMP, a hallmark of the Ih current, was abolished in neurons without PKA activity. Collectively, these results show a significant contribution of both genes to inflammatory pain and suggest that PKA-dependent activation of HCN2 underlies cAMP-triggered neuronal sensitization.
Collapse
|
131
|
O'Leary H, Smart KM, Moloney NA, Blake C, Doody CM. Pain sensitization associated with nonresponse after physiotherapy in people with knee osteoarthritis. Pain 2018; 159:1877-1886. [DOI: 10.1097/j.pain.0000000000001288] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
132
|
Sorkin LS, Eddinger KA, Woller SA, Yaksh TL. Origins of antidromic activity in sensory afferent fibers and neurogenic inflammation. Semin Immunopathol 2018; 40:237-247. [PMID: 29423889 PMCID: PMC7879713 DOI: 10.1007/s00281-017-0669-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/28/2017] [Indexed: 10/18/2022]
Abstract
Neurogenic inflammation results from the release of biologically active agents from the peripheral primary afferent terminal. This release reflects the presence of releasable pools of active product and depolarization-exocytotic coupling mechanisms in the distal afferent terminal and serves to alter the physiologic function of innervated organ systems ranging from the skin and meninges to muscle, bone, and viscera. Aside from direct stimulation, this biologically important release from the peripheral afferent terminal can be initiated by antidromic activity arising from five anatomically distinct points of origin: (i) afferent collaterals at the peripheral-target organ level, (ii) afferent collaterals arising proximal to the target organ, (iii) from mid-axon where afferents lacking myelin sheaths (C fibers and others following demyelinating injuries) may display crosstalk and respond to local irritation, (iv) the dorsal root ganglion itself, and (v) the central terminals of the afferent in the dorsal horn where local circuits and bulbospinal projections can initiate the so-called dorsal root reflexes, i.e., antidromic traffic in the sensory afferent.
Collapse
Affiliation(s)
- Linda S Sorkin
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA.
| | - Kelly A Eddinger
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA
| | - Sarah A Woller
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
133
|
Loos C, Moeller K, Fröhlich F, Hucho T, Hasenauer J. A Hierarchical, Data-Driven Approach to Modeling Single-Cell Populations Predicts Latent Causes of Cell-To-Cell Variability. Cell Syst 2018; 6:593-603.e13. [DOI: 10.1016/j.cels.2018.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/28/2017] [Accepted: 04/10/2018] [Indexed: 12/23/2022]
|
134
|
Hawkins JL, Moore NJ, Miley D, Durham PL. Secondary traumatic stress increases expression of proteins implicated in peripheral and central sensitization of trigeminal neurons. Brain Res 2018. [PMID: 29522721 DOI: 10.1016/j.brainres.2018.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The pathology of migraine, a common neurological disease, involves sensitization and activation of trigeminal nociceptive neurons to promote hyperalgesia and allodynia during an attack. Migraineurs often exhibit characteristics of a hyperexcitable or hypervigilant nervous system. One of the primary reported risk factors for development of a hyperexcitable trigeminal system is chronic, unmanaged stress and anxiety. While primary traumatic stress is a commonly cited risk factor for many pain conditions, exposure to secondary traumatic stress early in life is also thought to be a contributing risk factor. The goal of this study was to investigate cellular changes within the spinal trigeminal nucleus and trigeminal ganglion mediated by secondary traumatic stress. Male Sprague Dawley rats (sender) were subjected to forced swim testing (primary traumatic stress) and were then housed in close visual, olfactory, and auditory proximity to the breeding male and female rats, pregnant female rats, or female rats and their nursing offspring (all receivers). In response to secondary stress, levels of calcitonin gene-related peptide, active forms of the mitogen activated protein kinases ERK, JNK, and p38, and astrocyte expression of glial fibrillary acidic protein were significantly elevated in the spinal trigeminal nucleus in day 45 offspring when compared to naïve offspring. In addition, increased nuclear expression of ERK and p38 was observed in trigeminal ganglion neurons. Our results demonstrate that secondary traumatic stress promotes cellular events associated with prolonged trigeminal sensitization in the offspring, and provides a mechanism of how early life stress may function as a risk factor for migraine.
Collapse
Affiliation(s)
- J L Hawkins
- Missouri State University, JVIC-CBLS, 524 North Boonville Avenue, Springfield, MO 65806, United States
| | - N J Moore
- Missouri State University, JVIC-CBLS, 524 North Boonville Avenue, Springfield, MO 65806, United States
| | - D Miley
- Missouri State University, JVIC-CBLS, 524 North Boonville Avenue, Springfield, MO 65806, United States
| | - P L Durham
- Missouri State University, JVIC-CBLS, 524 North Boonville Avenue, Springfield, MO 65806, United States.
| |
Collapse
|
135
|
Antinociceptive Activity of Methanolic Extract of Clinacanthus nutans Leaves: Possible Mechanisms of Action Involved. Pain Res Manag 2018; 2018:9536406. [PMID: 29686743 PMCID: PMC5857305 DOI: 10.1155/2018/9536406] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023]
Abstract
Methanolic extract of Clinacanthus nutans Lindau leaves (MECN) has been proven to possess antinociceptive activity that works via the opioid and NO-dependent/cGMP-independent pathways. In the present study, we aimed to further determine the possible mechanisms of antinociception of MECN using various nociceptive assays. The antinociceptive activity of MECN was (i) tested against capsaicin-, glutamate-, phorbol 12-myristate 13-acetate-, bradykinin-induced nociception model; (ii) prechallenged against selective antagonist of opioid receptor subtypes (β-funaltrexamine, naltrindole, and nor-binaltorphimine); (iii) prechallenged against antagonist of nonopioid systems, namely, α2-noradrenergic (yohimbine), β-adrenergic (pindolol), adenosinergic (caffeine), dopaminergic (haloperidol), and cholinergic (atropine) receptors; (iv) prechallenged with inhibitors of various potassium channels (glibenclamide, apamin, charybdotoxin, and tetraethylammonium chloride). The results demonstrated that the orally administered MECN (100, 250, and 500 mg/kg) significantly (p < 0.05) reversed the nociceptive effect of all models in a dose-dependent manner. Moreover, the antinociceptive activity of 500 mg/kg MECN was significantly (p < 0.05) inhibited by (i) antagonists of μ-, δ-, and κ-opioid receptors; (ii) antagonists of α2-noradrenergic, β-adrenergic, adenosinergic, dopaminergic, and cholinergic receptors; and (iii) blockers of different K+ channels (voltage-activated-, Ca2+-activated, and ATP-sensitive-K+ channels, resp.). In conclusion, MECN-induced antinociception involves modulation of protein kinase C-, bradykinin-, TRVP1 receptors-, and glutamatergic-signaling pathways; opioidergic, α2-noradrenergic, β-adrenergic, adenosinergic, dopaminergic, and cholinergic receptors; and nonopioidergic receptors as well as the opening of various K+ channels. The antinociceptive activity could be associated with the presence of several flavonoid-based bioactive compounds and their synergistic action with nonvolatile bioactive compounds.
Collapse
|
136
|
Willemen HLDM, Kavelaars A, Prado J, Maas M, Versteeg S, Nellissen LJJ, Tromp J, Gonzalez Cano R, Zhou W, Jakobsson ME, Małecki J, Posthuma G, Habib AM, Heijnen CJ, Falnes PØ, Eijkelkamp N. Identification of FAM173B as a protein methyltransferase promoting chronic pain. PLoS Biol 2018; 16:e2003452. [PMID: 29444090 PMCID: PMC5828452 DOI: 10.1371/journal.pbio.2003452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 02/27/2018] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
Chronic pain is a debilitating problem, and insights in the neurobiology of chronic pain are needed for the development of novel pain therapies. A genome-wide association study implicated the 5p15.2 region in chronic widespread pain. This region includes the coding region for FAM173B, a functionally uncharacterized protein. We demonstrate here that FAM173B is a mitochondrial lysine methyltransferase that promotes chronic pain. Knockdown and sensory neuron overexpression strategies showed that FAM173B is involved in persistent inflammatory and neuropathic pain via a pathway dependent on its methyltransferase activity. FAM173B methyltransferase activity in sensory neurons hyperpolarized mitochondria and promoted macrophage/microglia activation through a reactive oxygen species–dependent pathway. In summary, we uncover a role for methyltransferase activity of FAM173B in the neurobiology of pain. These results also highlight FAM173B methyltransferase activity as a potential therapeutic target to treat debilitating chronic pain conditions. Pain is an evolutionarily conserved physiological phenomenon necessary for survival. Yet, pain can become pathological when it occurs independently of noxious stimuli. The molecular mechanisms of pathological pain are still poorly understood, limiting the development of highly needed novel analgesics. Recently, genetic variations in the genomic region encoding FAM173B—a functionally uncharacterized protein—have been linked to chronic pain in humans. In this study, we identify the role and function of FAM173B in the development of pathological pain. We used genetic, biochemical, and behavioral approaches in mice to show that FAM173B is a mitochondrial lysine methyltransferase—a protein that transfers methyl group to donor proteins. By genetically silencing or overexpressing FAM173B in sensory neurons, we showed that FAM173B methyltransferase activity promotes the development of chronic pain. In addition, we discovered that FAM173B methyltransferase activity in the mitochondria of sensory neurons promotes chronic pain via a pathway that depends on the production of reactive oxygen species and on the engagement of spinal cord microglia—engulfing cells of the central nervous system. These data point to an essential role of FAM173B in the regulation of pathological pain.
Collapse
Affiliation(s)
- Hanneke L. D. M. Willemen
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Annemieke Kavelaars
- Laboratory of Neuroimmunology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Judith Prado
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Mirjam Maas
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sabine Versteeg
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lara J. J. Nellissen
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jeshua Tromp
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rafael Gonzalez Cano
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Pharmacology and Institute of Neuroscience, University of Granada, Granada, Spain
| | - Wenjun Zhou
- Laboratory of Neuroimmunology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Magnus E. Jakobsson
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Jędrzej Małecki
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - George Posthuma
- Department of Cell Biology and Institute of Biomembranes, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Abdella M. Habib
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
- College of Medicine, Member of Qatar Health, Qatar University, Doha, Qatar
| | - Cobi J. Heijnen
- Laboratory of Neuroimmunology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Pål Ø. Falnes
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Niels Eijkelkamp
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
137
|
CD44 Signaling Mediates High Molecular Weight Hyaluronan-Induced Antihyperalgesia. J Neurosci 2017; 38:308-321. [PMID: 29175954 DOI: 10.1523/jneurosci.2695-17.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/08/2017] [Accepted: 11/13/2017] [Indexed: 01/12/2023] Open
Abstract
We studied, in male Sprague Dawley rats, the role of the cognate hyaluronan receptor, CD44 signaling in the antihyperalgesia induced by high molecular weight hyaluronan (HMWH). Low molecular weight hyaluronan (LMWH) acts at both peptidergic and nonpeptidergic nociceptors to induce mechanical hyperalgesia that is prevented by intrathecal oligodeoxynucleotide antisense to CD44 mRNA, which also prevents hyperalgesia induced by a CD44 receptor agonist, A6. Ongoing LMWH and A6 hyperalgesia are reversed by HMWH. HMWH also reverses the hyperalgesia induced by diverse pronociceptive mediators, prostaglandin E2, epinephrine, TNFα, and interleukin-6, and the neuropathic pain induced by the cancer chemotherapy paclitaxel. Although CD44 antisense has no effect on the hyperalgesia induced by inflammatory mediators or paclitaxel, it eliminates the antihyperalgesic effect of HMWH. HMWH also reverses the hyperalgesia induced by activation of intracellular second messengers, PKA and PKCε, indicating that HMWH-induced antihyperalgesia, although dependent on CD44, is mediated by an intracellular signaling pathway rather than as a competitive receptor antagonist. Sensitization of cultured small-diameter DRG neurons by prostaglandin E2 is also prevented and reversed by HMWH. These results demonstrate the central role of CD44 signaling in HMWH-induced antihyperalgesia, and establish it as a therapeutic target against inflammatory and neuropathic pain.SIGNIFICANCE STATEMENT We demonstrate that hyaluronan (HA) with different molecular weights produces opposing nociceptive effects. While low molecular weight HA increases sensitivity to mechanical stimulation, high molecular weight HA reduces sensitization, attenuating inflammatory and neuropathic hyperalgesia. Both pronociceptive and antinociceptive effects of HA are mediated by activation of signaling pathways downstream CD44, the cognate HA receptor, in nociceptors. These results contribute to our understanding of the role of the extracellular matrix in pain, and indicate CD44 as a potential therapeutic target to alleviate inflammatory and neuropathic pain.
Collapse
|
138
|
Payrits M, Sághy É, Cseko K, Pohóczky K, Bölcskei K, Ernszt D, Barabás K, Szolcsányi J, Ábrahám IM, Helyes Z, Szoke É. Estradiol Sensitizes the Transient Receptor Potential Vanilloid 1 Receptor in Pain Responses. Endocrinology 2017; 158:3249-3258. [PMID: 28977586 DOI: 10.1210/en.2017-00101] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/29/2017] [Indexed: 11/19/2022]
Abstract
Sex differences exist in chronic pain pathologies, and gonadal estradiol (E2) alters the pain sensation. The nocisensor transient receptor potential vanilloid 1 (TRPV1) receptor plays a critical role in triggering pain. Here we examined the impact of E2 on the function of TRPV1 receptor in mice sensory neurons in vitro and in vivo. Both mechano- and thermonociceptive thresholds of the plantar surface of the paw of female mice were significantly lower in proestrus compared with the estrus phase. These thresholds were higher in ovariectomized (OVX) mice and significantly lower in sham-operated mice in proestrus compared with the sham-operated mice in estrus phase. This difference was absent in TRPV1 receptor-deficient mice. Furthermore, E2 potentiated the TRPV1 receptor activation-induced mechanical hyperalgesia in OVX mice. Long pretreatment (14 hours) with E2 induced a significant increase in TRPV1 receptor messenger RNA expression and abolished the capsaicin-induced TRPV1 receptor desensitization in primary sensory neurons. The short E2 incubation (10 minutes) also prevented the desensitization, which reverted after coadministration of E2 and the tropomyosin-related kinase A (TrkA) receptor inhibitor. Our study provides in vivo and in vitro evidence for E2-induced TRPV1 receptor upregulation and sensitization mediated by TrkAR via E2-induced genomic and nongenomic mechanisms. The sensitization and upregulation of TRPV1 receptor by E2 in sensory neurons may explain the greater pain sensitivity in female mice.
Collapse
Affiliation(s)
- Maja Payrits
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Éva Sághy
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089, Budapest, Hungary
| | - Kata Cseko
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Krisztina Pohóczky
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Dávid Ernszt
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- Department of Pharmaceutical Biotechnology, University of Pécs, Medical School, H-7624 Pécs, Hungary
| | - Klaudia Barabás
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- Institute of Physiology, University of Pécs, Medical School, H-7624 Pécs, Hungary
| | - János Szolcsányi
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
| | - István M Ábrahám
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- Institute of Physiology, University of Pécs, Medical School, H-7624 Pécs, Hungary
- MTA-PTE NAP B Molecular Neuroendocrinology Research Group-Hungary, H-7624 Pécs, Hungary
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP B Chronic Pain Research Group-Hungary, H-7624 Pécs, Hungary
| | - Éva Szoke
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP B Chronic Pain Research Group-Hungary, H-7624 Pécs, Hungary
| |
Collapse
|
139
|
Jiang L, Pan CL, Wang CY, Liu BQ, Han Y, Hu L, Liu L, Yang Y, Qu JW, Liu WT. Selective suppression of the JNK-MMP2/9 signal pathway by tetramethylpyrazine attenuates neuropathic pain in rats. J Neuroinflammation 2017; 14:174. [PMID: 28859670 PMCID: PMC5580313 DOI: 10.1186/s12974-017-0947-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/23/2017] [Indexed: 12/13/2022] Open
Abstract
Background Activated astrocytes release matrix metalloproteinase-2/9 (MMP-2/9) to induce central sensitization and maintain neuropathic pain. However, the mechanisms involved in the activation of MMP-2/9 on astrocytes during pain remain poorly understood. Meanwhile, there is a lack of effective treatment to inhibit the activation of MMP-2/9 on astrocytes. In this study, we aim to investigate the effect of tetramethylpyrazine (TMP), a natural compound with analgesic effects but unknown mechanisms, on MMP-2/9 in neuropathic pain. Methods The nociception was assessed by measuring the incidence of foot withdrawal in response to mechanical indentation in rats (n = 6). Cell signaling was assayed using western blotting (n = 6) and immunohistochemistry (n = 5). The astrocyte cell line C8-D1A was cultured to investigate the in vitro effects. Results TMP significantly attenuated the maintenance of chronic constrictive injury (CCI)-induced neuropathic pain, inhibited the activation of astrocytes, and decreased the expression of MMP-2/9. Furthermore, our results indicated that TMP could selectively suppress JNK activity but had no notable effects on ERK and p38. Our study also revealed that the effect of TMP may be dependent on the inhibition of TAK1. Conclusions Inhibition of astrocyte activation in the spinal cord by tetramethylpyrazine may have utility in the treatment of CCI-induced neuroinflammation, and our results further implicate JNK-MMP-2/9 as a novel target for the attenuation of neuropathic pain. Electronic supplementary material The online version of this article (10.1186/s12974-017-0947-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lai Jiang
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Han-Zhong Road, Nanjing, 210029, China
| | - Cai-Long Pan
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Han-Zhong Road, Nanjing, 210029, China
| | - Chao-Yu Wang
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Han-Zhong Road, Nanjing, 210029, China
| | - Bing-Qian Liu
- Department of Ophthalmology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Yuan Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, 221000, China
| | - Liang Hu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Han-Zhong Road, Nanjing, 210029, China
| | - Lei Liu
- Department of pain, Shandong Qianfoshan Hospital, Shandong, 250014, China
| | - Yang Yang
- Department of Gynecologic Oncology, Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, 210009, China
| | - Jun-Wei Qu
- Department of Gynecologic Oncology, Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, 210009, China
| | - Wen-Tao Liu
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 140 Han-Zhong Road, Nanjing, 210029, China.
| |
Collapse
|
140
|
de Araújo JCB, Gondim DV, Cavalcante ALC, Lisboa MRP, de Castro Brito GA, Vale ML. Inflammatory pain assessment in the arthritis of the temporomandibular joint in rats: A comparison between two phlogistic agents. J Pharmacol Toxicol Methods 2017; 88:100-108. [PMID: 28797764 DOI: 10.1016/j.vascn.2017.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 07/21/2017] [Accepted: 08/05/2017] [Indexed: 12/01/2022]
Abstract
Temporomandibular joint (TMJ) disorders are a group of conditions that result in TMJ pain, which frequently limits basic daily activities. Experimental models that allow the study of the mechanisms underlying these inflammatory and pain conditions are of great clinical relevance. The aim of this study was to evaluate nociception, inflammation and participation of the macrophage/microglia cells in the arthritis of the TMJ induced by two phlogistic agents. 84 rats were divided into 2 groups: Zy, which received zymosan intra-articularly, or Cg, which received carrageenan intra-articularly. Mechanical nociception, total leukocyte influx to the synovial fluid and histopathological analyses were evaluated in the TMJ. The participation of macrophage/microglia located in trigeminal ganglia (TG) and in the subnucleus caudalis (V-SnC) was assessed immunohistochemically. Both agents induced mechanical hyperalgesia 6h after the induction, but a more persistent algesic state was perceived in the Cg group, which lasted for 120h. Even though both groups presented increased leukocyte influx, the Zy-group presented a more intense influx. Zymosan recruited resident macrophage in the trigeminal ganglia 24h after the injection. In the V-SnC, the group Cg presented a more prolonged immunolabeling pattern in comparison with the group Zy. It can be concluded that zymosan induced a more intense infiltrate and peripheral nervous changes, while Cg lead to a moderate TMJ inflammation with prominent changes in the V-SnC.
Collapse
Affiliation(s)
| | - Delane Viana Gondim
- Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil
| | - André Luiz Cunha Cavalcante
- Medical Sciences Post Graduation Program, Department of Clinical Medicine, Federal University of Ceará, Brazil
| | - Mario Roberto Pontes Lisboa
- Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil
| | - Gerly Anne de Castro Brito
- Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil
| | - Mariana Lima Vale
- Pharmacology Post Graduation Program, Department of Physiology and Pharmacology, Federal University of Ceará, Brazil; Morphofunctional Sciences Post Graduation Program, Department of Morphology, Federal University of Ceará, Brazil.
| |
Collapse
|
141
|
Singh H, Bhushan S, Arora R, Singh Buttar H, Arora S, Singh B. Alternative treatment strategies for neuropathic pain: Role of Indian medicinal plants and compounds of plant origin-A review. Biomed Pharmacother 2017; 92:634-650. [DOI: 10.1016/j.biopha.2017.05.079] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 01/28/2023] Open
|
142
|
Calcineurin Dysregulation Underlies Spinal Cord Injury-Induced K + Channel Dysfunction in DRG Neurons. J Neurosci 2017; 37:8256-8272. [PMID: 28751455 DOI: 10.1523/jneurosci.0434-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/26/2022] Open
Abstract
Dysfunction of the fast-inactivating Kv3.4 potassium current in dorsal root ganglion (DRG) neurons contributes to the hyperexcitability associated with persistent pain induced by spinal cord injury (SCI). However, the underlying mechanism is not known. In light of our previous work demonstrating modulation of the Kv3.4 channel by phosphorylation, we investigated the role of the phosphatase calcineurin (CaN) using electrophysiological, molecular, and imaging approaches in adult female Sprague Dawley rats. Pharmacological inhibition of CaN in small-diameter DRG neurons slowed repolarization of the somatic action potential (AP) and attenuated the Kv3.4 current. Attenuated Kv3.4 currents also exhibited slowed inactivation. We observed similar effects on the recombinant Kv3.4 channel heterologously expressed in Chinese hamster ovary cells, supporting our findings in DRG neurons. Elucidating the molecular basis of these effects, mutation of four previously characterized serines within the Kv3.4 N-terminal inactivation domain eliminated the effects of CaN inhibition on the Kv3.4 current. SCI similarly induced concurrent Kv3.4 current attenuation and slowing of inactivation. Although there was little change in CaN expression and localization after injury, SCI induced upregulation of the native regulator of CaN 1 (RCAN1) in the DRG at the transcript and protein levels. Consistent with CaN inhibition resulting from RCAN1 upregulation, overexpression of RCAN1 in naive DRG neurons recapitulated the effects of pharmacological CaN inhibition on the Kv3.4 current and the AP. Overall, these results demonstrate a novel regulatory pathway that links CaN, RCAN1, and Kv3.4 in DRG neurons. Dysregulation of this pathway might underlie a peripheral mechanism of pain sensitization induced by SCI.SIGNIFICANCE STATEMENT Pain sensitization associated with spinal cord injury (SCI) involves poorly understood maladaptive modulation of neuronal excitability. Although central mechanisms have received significant attention, recent studies have identified peripheral nerve hyperexcitability as a driver of persistent pain signaling after SCI. However, the ion channels and signaling molecules responsible for this change in primary sensory neuron excitability are still not well defined. To address this problem, this study used complementary electrophysiological and molecular methods to determine how Kv3.4, a voltage-gated K+ channel robustly expressed in dorsal root ganglion neurons, becomes dysfunctional upon calcineurin (CaN) inhibition. The results strongly suggest that CaN inhibition underlies SCI-induced dysfunction of Kv3.4 and the associated excitability changes through upregulation of the native regulator of CaN 1 (RCAN1).
Collapse
|
143
|
Abstract
Pain associated with mechanical, chemical, and thermal heat stimulation of the ocular surface is mediated by trigeminal ganglion neurons, while cold thermoreceptors detect wetness and reflexly maintain basal tear production and blinking rate. These neurons project into two regions of the trigeminal brain stem nuclear complex: ViVc, activated by changes in the moisture of the ocular surface and VcC1, mediating sensory-discriminative aspects of ocular pain and reflex blinking. ViVc ocular neurons project to brain regions that control lacrimation and spontaneous blinking and to the sensory thalamus. Secretion of the main lacrimal gland is regulated dominantly by autonomic parasympathetic nerves, reflexly activated by eye surface sensory nerves. These also evoke goblet cell secretion through unidentified efferent fibers. Neural pathways involved in the regulation of meibomian gland secretion or mucin release have not been identified. In dry eye disease, reduced tear secretion leads to inflammation and peripheral nerve damage. Inflammation causes sensitization of polymodal and mechano-nociceptor nerve endings and an abnormal increase in cold thermoreceptor activity, altogether evoking dryness sensations and pain. Long-term inflammation and nerve injury alter gene expression of ion channels and receptors at terminals and cell bodies of trigeminal ganglion and brainstem neurons, changing their excitability, connectivity and impulse firing. Perpetuation of molecular, structural and functional disturbances in ocular sensory pathways ultimately leads to dysestesias and neuropathic pain referred to the eye surface. Pain can be assessed with a variety of questionaires while the status of corneal nerves is evaluated with esthesiometry and with in vivo confocal microscopy.
Collapse
|
144
|
Goldstein RH, Katz B, Lev S, Binshtok AM. Ultrafast optical recording reveals distinct capsaicin-induced ion dynamics along single nociceptive neurite terminals in vitro. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:76010. [PMID: 28715544 DOI: 10.1117/1.jbo.22.7.076010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/26/2017] [Indexed: 06/07/2023]
Abstract
Pain signals are detected by terminals of nociceptive peripheral fibers situated among the keratinocytes and epithelial cells. Despite being key structures for pain-related stimuli detection and transmission, little is known about the functional organization of terminals. This is mainly due to their minute size, rendering them largely inaccessible by conventional experimental approaches. Here, we report the implementation of an ultrafast optical recording approach for studying cultured neurite terminals, which are readily accessible for assay manipulations. Using this approach, we were able to study capsaicin-induced calcium and sodium dynamics in the nociceptive processes, at a near-action potential time resolution. The approach was sensitive enough to detect differences in latency, time-to-peak, and amplitude of capsaicin-induced ion transients along the terminal neurites. Using this approach, we found that capsaicin evokes distinctive calcium signals along the neurite. At the terminal, the signal was insensitive to voltage-gated sodium channel blockers, and showed slower kinetics and smaller signal amplitudes, compared with signals that were measured further up the neurite. These latter signals were mainly abolished by sodium channel blockers. We propose this ultrafast optical recording approach as a model for studying peripheral terminal signaling, forming a basis for studying pain mechanisms in normal and pathological states.
Collapse
Affiliation(s)
- Robert H Goldstein
- The Hebrew University, Institute for Medical Research Israel Canada, Faculty of Medicine, Department of Medical Neurobiology, Jerusalem, IsraelbThe Hebrew University, The Edmond and Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Ben Katz
- The Hebrew University, Institute for Medical Research Israel Canada, Faculty of Medicine, Department of Medical Neurobiology, Jerusalem, IsraelbThe Hebrew University, The Edmond and Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Shaya Lev
- The Hebrew University, Institute for Medical Research Israel Canada, Faculty of Medicine, Department of Medical Neurobiology, Jerusalem, IsraelbThe Hebrew University, The Edmond and Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Alexander M Binshtok
- The Hebrew University, Institute for Medical Research Israel Canada, Faculty of Medicine, Department of Medical Neurobiology, Jerusalem, IsraelbThe Hebrew University, The Edmond and Lily Safra Center for Brain Sciences, Jerusalem, Israel
| |
Collapse
|
145
|
Fischer BD, Ho C, Kuzin I, Bottaro A, O’Leary ME. Chronic exposure to tumor necrosis factor in vivo induces hyperalgesia, upregulates sodium channel gene expression and alters the cellular electrophysiology of dorsal root ganglion neurons. Neurosci Lett 2017; 653:195-201. [DOI: 10.1016/j.neulet.2017.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/01/2017] [Accepted: 05/03/2017] [Indexed: 12/25/2022]
|
146
|
Chen G, Kim YH, Li H, Luo H, Liu DL, Zhang ZJ, Lay M, Chang W, Zhang YQ, Ji RR. PD-L1 inhibits acute and chronic pain by suppressing nociceptive neuron activity via PD-1. Nat Neurosci 2017; 20:917-926. [PMID: 28530662 PMCID: PMC5831162 DOI: 10.1038/nn.4571] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/27/2017] [Indexed: 12/14/2022]
Abstract
Programmed cell death ligand-1 (PD-L1) is typically produced by cancer cells and suppresses immunity through the receptor PD-1 expressed on T cells. However, the role of PD-L1 and PD-1 in regulating pain and neuronal function is unclear. Here we report that both melanoma and normal neural tissues including dorsal root ganglion (DRG) produce PD-L1 that can potently inhibit acute and chronic pain. Intraplantar injection of PD-L1 evoked analgesia in naive mice via PD-1, whereas PD-L1 neutralization or PD-1 blockade induced mechanical allodynia. Mice lacking Pd1 (Pdcd1) exhibited thermal and mechanical hypersensitivity. PD-1 activation in DRG nociceptive neurons by PD-L1 induced phosphorylation of the tyrosine phosphatase SHP-1, inhibited sodium channels and caused hyperpolarization through activation of TREK2 K+ channels. PD-L1 also potently suppressed nociceptive neuron excitability in human DRGs. Notably, blocking PD-L1 or PD-1 elicited spontaneous pain and allodynia in melanoma-bearing mice. Our findings identify a previously unrecognized role of PD-L1 as an endogenous pain inhibitor and a neuromodulator.
Collapse
Affiliation(s)
- Gang Chen
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
- Department of Physiology, College of Medicine, Gachon University, Incheon, Korea
| | - Hui Li
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Hao Luo
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Da-Lu Liu
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Zhi-Jun Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Mark Lay
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Yu-Qiu Zhang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710
| |
Collapse
|
147
|
Shah M, Choi S. Toll-like Receptor-Dependent Negative Effects of Opioids: A Battle between Analgesia and Hyperalgesia. Front Immunol 2017; 8:642. [PMID: 28620391 PMCID: PMC5450035 DOI: 10.3389/fimmu.2017.00642] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/17/2017] [Indexed: 11/28/2022] Open
Abstract
Our understanding of the pathophysiology of the pathological pain and the pharmacology of analgesic treatments has progressed tremendously over the past two decades. Among the well-documented pro-algesic factors, glia and other toll-like receptors (TLRs)-expressing cells in the neuroimmune interface have been recognized for their role in the development of neuropathic pain and for compromising the analgesic effects of opioids. Here, we comprehensively review the molecular mechanisms of pain initiation and progression, the role of TLRs in these processes, and the molecular mechanisms of morphine and morphine-3-glucuronide in TLR-dependent central immune signaling. The data reviewed here suggest that, while targeting glia to treat neuropathic pain, both analgesic and analgesia-opposing effects of opioids must be considered by acknowledging their role in TLR-mediated signaling.
Collapse
Affiliation(s)
- Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
148
|
Isensee J, Schild C, Schwede F, Hucho T. Crosstalk from cAMP to ERK1/2 emerges during postnatal maturation of nociceptive neurons and is maintained during aging. J Cell Sci 2017; 130:2134-2146. [PMID: 28515230 DOI: 10.1242/jcs.197327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 05/10/2017] [Indexed: 02/02/2023] Open
Abstract
Maturation of nociceptive neurons depends on changes in transcription factors, ion channels and neuropeptides. Mature nociceptors initiate pain in part by drastically reducing the activation threshold via intracellular sensitization signaling. Whether sensitization signaling also changes during development and aging remains so far unknown. Using a novel automated microscopy approach, we quantified changes in intracellular signaling protein expression and in their signaling dynamics, as well as changes in intracellular signaling cascade wiring, in sensory neurons from newborn to senescent (24 months of age) rats. We found that nociceptive subgroups defined by the signaling components protein kinase A (PKA)-RIIβ (also known as PRKAR2B) and CaMKIIα (also known as CAMK2A) developed at around postnatal day 10, the time of nociceptor maturation. The integrative nociceptor marker, PKA-RIIβ, allowed subgroup segregation earlier than could be achieved by assessing the classical markers TRPV1 and Nav1.8 (also known as SCN10A). Signaling kinetics remained constant over lifetime despite in part strong changes in the expression levels. Strikingly, we found a mechanism important for neuronal memory - i.e. the crosstalk from cAMP and PKA to ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1, respectively) - to emerge postnatally. Thus, maturation of nociceptors is closely accompanied by altered expression, activation and connectivity of signaling pathways known to be central for pain sensitization and neuronal memory formation.
Collapse
Affiliation(s)
- Joerg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Robert Koch Str. 10, Cologne D-50931, Germany
| | - Cosimo Schild
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Robert Koch Str. 10, Cologne D-50931, Germany
| | - Frank Schwede
- BIOLOG Life Science Institute, Flughafendamm 9A, Bremen D-28199, Germany
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Robert Koch Str. 10, Cologne D-50931, Germany
| |
Collapse
|
149
|
Tabuena DR, Solis A, Geraldi K, Moffatt CA, Fuse M. Central neural alterations predominate in an insect model of nociceptive sensitization. J Comp Neurol 2017; 525:1176-1191. [PMID: 27650422 PMCID: PMC5258852 DOI: 10.1002/cne.24124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/20/2016] [Accepted: 09/11/2016] [Indexed: 12/12/2022]
Abstract
Many organisms respond to noxious stimuli with defensive maneuvers. This is noted in the hornworm, Manduca sexta, as a defensive strike response. After tissue damage, organisms typically display sensitized responses to both noxious or normally innocuous stimuli. To further understand this phenomenon, we used novel in situ and in vitro preparations based on paired extracellular nerve recordings and videography to identify central and peripheral nerves responsible for nociception and sensitization of the defensive behavior in M. sexta. In addition, we used the in vivo defensive strike response threshold assayed with von Frey filaments to examine the roles that N-methyl-D-aspartate receptor (NMDAR) and hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels play in this nociceptive sensitization using the inhibitors MK-801 and AP5 (NMDAR), and ivabradine and ZD7288 (HCN). Using our new preparations, we found that afferent activity evoked by noxious pinch in these preparations was conveyed to central ganglia by axons in the anterior- and lateral-dorsal nerve branches, and that sensitization induced by tissue damage was mediated centrally. Furthermore, sensitization was blocked by all inhibitors tested except the inactive isomer L-AP5, and reversed by ivabradine both in vivo and in vitro. Our findings suggest that M. sexta's sensitization occurs through central signal amplification. Due to the relatively natural sensitization method and conserved molecular actions, we suggest that M. sexta may be a valuable model for studying the electrophysiological properties of nociceptive sensitization and potentially related conditions such as allodynia and hyperalgesia in a comparative setting that offers unique experimental advantages. J. Comp. Neurol. 525:1176-1191, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dennis R Tabuena
- San Francisco State University, Dept. Biology. 1600 Holloway Avenue, San Francisco, CA 94132
| | - Allan Solis
- City College of San Francisco, 50 Phelan Ave, San Francisco, CA 94112
| | - Ken Geraldi
- San Francisco State University, Dept. Biology. 1600 Holloway Avenue, San Francisco, CA 94132
| | - Christopher A Moffatt
- San Francisco State University, Dept. Biology. 1600 Holloway Avenue, San Francisco, CA 94132
| | - Megumi Fuse
- San Francisco State University, Dept. Biology. 1600 Holloway Avenue, San Francisco, CA 94132
| |
Collapse
|
150
|
Abstract
The treatment of neuropathic pain remains a clinical challenge because of its unclear mechanisms and broad clinical morbidity. Matrix metalloproteinase (MMP)-9 and MMP-2 have previously been described as key components in neuropathic pain because of their facilitation of inflammatory cytokine maturation and induction of neural inflammation. Therefore, the inhibition of MMPs may represent a novel therapeutic approach to the treatment of neuropathic pain. In this study, we report that N-acetyl-cysteine (NAC), which is a broadly used respiratory drug, significantly attenuates neuropathic pain through a unique mechanism of MMP inhibition. Both the in vitro (0.1 mM) and in vivo application of NAC significantly suppressed the activity of MMP-9/2. Orally administered NAC (50, 100, and 200 mg/kg) not only postponed the occurrence but also inhibited the maintenance of chronic constrictive injury (CCI)-induced neuropathic pain in rats. The administration of NAC blocked the maturation of interleukin-1β, which is a critical substrate of MMPs, and markedly suppressed the neuronal activation induced by CCI, including inhibiting the phosphorylation of protein kinase Cγ, NMDAR1, and mitogen-activated protein kinases. Finally, NAC significantly inhibited CCI-induced microglia activation but elicited no notable effects on astrocytes. These results demonstrate an effective and safe approach that has been used clinically to alleviate neuropathic pain through the powerful inhibition of the activation of MMPs.
Collapse
|