51
|
Sustained and repeated mouth opening leads to development of painful temporomandibular disorders involving macrophage/microglia activation in mice. Pain 2019. [PMID: 29533386 DOI: 10.1097/j.pain.0000000000001206] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Temporomandibular disorder (TMD) is a set of heterogeneous musculoskeletal conditions involving the temporomandibular joint (TMJ) and/or the masticatory muscles. Up to 33% of the population has had at least 1 symptom of TMD with 5% to 10% of them requiring treatment. Common symptoms include limited jaw movement, joint sound, and pain in the orofacial area. Once TMD becomes chronic, it can be debilitating with comorbidities that greatly reduce one's overall quality of life. However, the underlying mechanism of TMD is unclear because of the multicausative nature of the disease. Here, we report a novel mouse model of TMD where a bite block was placed in between the upper and lower incisors such that the mouth was kept maximally open for 1.5 hours per day for 5 days. After sustained mouth opening, mice developed persistent orofacial mechanical allodynia and TMJ dysfunction. At the cellular level, we found masseter muscle dystrophy, and increased proteoglycan deposition and hypertrophic chondrocytes in the mandibular condyle. Increased F4/80 macrophages were also observed in the masseter muscles and the TMJ posterior synovium. We also found ATF3 neuronal injury and increased F4/80 macrophages in the trigeminal ganglia. Microglia activation was observed in the trigeminal subnucleus caudalis. Inhibiting macrophage and microglia activation with a colony stimulating factor-1 receptor inhibitor prevented the development of orofacial mechanical allodynia, but not TMJ dysfunction. This study suggests that mouth opening for an extended period during dental treatments or oral intubations may risk the development of chronic TMD and inflammation associated with macrophage and microglia in the tissue and trigeminal system contributes to the development of TMD pain.
Collapse
|
52
|
Li Z, Wu F, Xu D, Zhi Z, Xu G. Inhibition of TREM1 reduces inflammation and oxidative stress after spinal cord injury (SCI) associated with HO-1 expressions. Biomed Pharmacother 2019; 109:2014-2021. [DOI: 10.1016/j.biopha.2018.08.159] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/26/2022] Open
|
53
|
Liu Z, Chen S, Qiu C, Sun Y, Li W, Jiang J, Zhang JM. Fractalkine/CX3CR1 Contributes to Endometriosis-Induced Neuropathic Pain and Mechanical Hypersensitivity in Rats. Front Cell Neurosci 2018; 12:495. [PMID: 30622457 PMCID: PMC6309014 DOI: 10.3389/fncel.2018.00495] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 12/03/2018] [Indexed: 11/13/2022] Open
Abstract
Pain is the most severe and common symptom of endometriosis. Its underlying pathogenetic mechanism is poorly understood. Nerve sensitization is a particular research challenge, due to the limitations of general endometriosis models and sampling nerve tissue from patients. The chemokine fractalkine (FKN) has been demonstrated to play a key role in various forms of neuropathic pain, while its role in endometriotic pain is unknown. Our study was designed to explore the function of FKN in the development and maintenance of peripheral hyperalgesia and central sensitization in endometriosis using a novel endometriosis animal model developed in our laboratory. After modeling, behavioral tests were carried out and the optimal time for molecular changes was obtained. We extracted ectopic tissues and L4-6 spinal cords to detect peripheral and central roles for FKN, respectively. To assess morphologic characteristics of endometriosis-like lesions-as well as expression and location of FKN/CX3CR1-we performed H&E staining, immunostaining, and western blotting analyses. Furthermore, inhibition of FKN expression in the spinal cord was achieved by intrathecal administration of an FKN-neutralizing antibody to demonstrate its function. Our results showed that implanted autologous uterine tissue around the sciatic nerve induced endometriosis-like lesions and produced mechanical hyperalgesia and allodynia. FKN was highly expressed on macrophages, whereas its receptor CX3CR1 was overexpressed in the myelin sheath of sciatic nerve fibers. Overexpressed FKN was also observed in neurons. CX3CR1/pp38-MAPK was upregulated in activated microglia in the spinal dorsal horn. Intrathecal administration of FKN-neutralizing antibody not only reversed the established mechanical hyperalgesia and allodynia, but also inhibited the expression of CX3CR1/pp38-MAPK in activated microglia, which was essential for the persistence of central sensitization. We concluded that the FKN/CX3CR1 signaling pathway might be one of the mechanisms of peripheral hyperalgesia in endometriosis, which requires further studies. Spinal FKN is important for the development and maintenance of central sensitization in endometriosis, and it may further serve as a novel therapeutic target to relieve persistent pain associated with endometriosis.
Collapse
Affiliation(s)
- Zhiming Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecologic Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Sisi Chen
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Chunping Qiu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecologic Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Yaqiong Sun
- Department of Obstetrics and Gynecology, Shandong Obstetrics and Gynecology Hospital, Jinan, China
| | - Wenzhi Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Gynecologic Oncology of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
54
|
Huang X, Wang W, Liu X, Xi Y, Yu J, Yang X, Ye X. Bone mesenchymal stem cells attenuate radicular pain by inhibiting microglial activation in a rat noncompressive disk herniation model. Cell Tissue Res 2018; 374:99-110. [PMID: 29858667 DOI: 10.1007/s00441-018-2855-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/08/2018] [Indexed: 12/25/2022]
Abstract
Spinal disk herniation can induce radicular pain through chemical irritation caused by proinflammatory and immune responses. Bone marrow mesenchymal stem cells (BMSCs) are a unique type of adult stem cell with the functions of suppressing inflammation and modulating immune responses. This study was undertaken to observe the effect of intrathecal BMSCs on the treatment of mechanical allodynia and the suppression of microglial activation in a rat noncompressive disk herniation model. The model was induced by the application of nucleus pulposus (NP) to the L5 dorsal root ganglion (DRG). The study found that the use of NP in the DRG can induce abnormal mechanical pain, increase the contents of the proinflammatory factors TNF-α and IL-1β, decrease the content of the anti-inflammatory cytokine TGF-β1 and activate microglia in the spinal dorsal horns (L5) (P < 0.05). BMSC administration could increase the mechanical withdrawal thresholds dramatically, decrease the contents of IL-1β and TNF-α, increase the content of TGF-β1 significantly (P < 0.05) and inhibit microglial activation in the bilateral spinal dorsal horn. Our results indicate that BMSC administration can reduce mechanical allodynia and downregulate the expression of proinflammatory cytokines by inhibiting microglial activation in the spinal dorsal horn in a rat noncompressive disk herniation model.
Collapse
Affiliation(s)
- Xiaodong Huang
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Weiheng Wang
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Xilin Liu
- Department of Orthopaedics, Chengdu General Hospital of Chengdu Military Command Region, Chengdu, 610083, China
| | - Yanhai Xi
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Jiangming Yu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Xiangqun Yang
- Department of Anatomy, Institute of Biomedical Engineering, Second Military Medical University, Shanghai, 200433, China.
| | - Xiaojian Ye
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| |
Collapse
|
55
|
Aman Y, Pitcher T, Ballard C, Malcangio M. Impaired chronic pain-like behaviour and altered opioidergic system in the TASTPM mouse model of Alzheimer's disease. Eur J Pain 2018; 23:91-106. [PMID: 29987897 PMCID: PMC6492091 DOI: 10.1002/ejp.1288] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2018] [Indexed: 11/08/2022]
Abstract
BACKGROUND Chronic pain conditions, especially osteoarthritis (OA), are as common in individuals with Alzheimer's disease (AD) as in the general elderly population, which results in detrimental impact on patient's quality of life. However, alteration in perception of pain in AD coupled with deteriorating ability to communicate pain sensations often result in under-diagnosis and inappropriate management of pain. Therefore, a better understanding of mechanisms in chronic pain processing in AD is needed. Here, we explored the development and progression of OA pain and the effect of analgesics in a transgenic mouse model of AD. METHODS Unilateral OA pain was induced chemically, via an intra-articular injection of monosodium iodoacetate (MIA) in the left knee joint of AD-mice (TASTPM) and age- and gender-matched C57BL/6J (WT). Pharmacological and biochemical assessments were conducted in plasma and spinal cord tissue. RESULTS MIA resulted in hind paw mechanical hypersensitivity (allodynia), initiating on day 3, in TASTPM and WT controls. However, from 14 to 28 days, TASTPM displayed partial attenuation of allodynia and diminished spinal microglial response compared to WT controls. Naloxone, an opioid antagonist, re-established allodynia levels as observed in the WT group. Morphine, an opioid agonist, induced heightened analgesia in AD-mice whilst gabapentin was devoid of efficacy. TASTPM exhibited elevated plasma level of β-endorphin post-MIA which correlated with impaired allodynia. CONCLUSIONS These results indicate an alteration of the opioidergic system in TASTPM as possible mechanisms underlying impaired persistent pain sensitivity in AD. This work provides basis for re-evaluation of opioid analgesic use for management of pain in AD. SIGNIFICANCE This study shows attenuated pain-like behaviour in a transgenic mouse model of Alzheimer's disease due to alterations in the opioidergic system and central plasticity mechanisms of persistent pain.
Collapse
Affiliation(s)
- Y Aman
- Wolfson Centre for Age Related Diseases, King's College London, UK
| | - T Pitcher
- Wolfson Centre for Age Related Diseases, King's College London, UK
| | - C Ballard
- Wolfson Centre for Age Related Diseases, King's College London, UK.,Medical School, University of Exeter, UK
| | - M Malcangio
- Wolfson Centre for Age Related Diseases, King's College London, UK
| |
Collapse
|
56
|
Albrecht D, Ahmed S, Kettner N, Borra R, Cohen-Adad J, Deng H, Houle T, Opalacz A, Roth S, Melo MV, Chen L, Mao J, Hooker J, Loggia ML, Zhang Y. Neuroinflammation of the spinal cord and nerve roots in chronic radicular pain patients. Pain 2018; 159:968-977. [PMID: 29419657 PMCID: PMC5908728 DOI: 10.1097/j.pain.0000000000001171] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Numerous preclinical studies support the role of spinal neuroimmune activation in the pathogenesis of chronic pain, and targeting glia (eg, microglia/astrocyte)- or macrophage-mediated neuroinflammatory responses effectively prevents or reverses the establishment of persistent nocifensive behaviors in laboratory animals. However, thus far, the translation of those findings into novel treatments for clinical use has been hindered by the scarcity of data supporting the role of neuroinflammation in human pain. Here, we show that patients suffering from a common chronic pain disorder (lumbar radiculopathy), compared with healthy volunteers, exhibit elevated levels of the neuroinflammation marker 18 kDa translocator protein, in both the neuroforamina (containing dorsal root ganglion and nerve roots) and spinal cord. These elevations demonstrated a pattern of spatial specificity correlating with the patients' clinical presentation, as they were observed in the neuroforamen ipsilateral to the symptomatic leg (compared with both contralateral neuroforamen in the same patients as well as to healthy controls) and in the most caudal spinal cord segments, which are known to process sensory information from the lumbosacral nerve roots affected in these patients (compared with more superior segments). Furthermore, the neuroforaminal translocator protein signal was associated with responses to fluoroscopy-guided epidural steroid injections, supporting its role as an imaging marker of neuroinflammation, and highlighting the clinical significance of these observations. These results implicate immunoactivation at multiple levels of the nervous system as a potentially important and clinically relevant mechanism in human radicular pain, and suggest that therapies targeting immune cell activation may be beneficial for chronic pain patients.
Collapse
Affiliation(s)
- Daniel Albrecht
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114
| | - Shihab Ahmed
- MGH Translational Pain Research Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Norman Kettner
- Department of Radiology, Logan University, Chesterfield, MO, 63017
| | - Ronald Borra
- Medical Imaging Centre of Southwest Finland, Turku University Hospital, Turku, Finland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Julien Cohen-Adad
- Department of Electrical Engineering, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Quebec, Canada; Functional Neuroimaging Unit, CRIUGM, Université de Montréal, Montreal, Quebec, Canada
| | - Hao Deng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Timothy Houle
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Arissa Opalacz
- MGH Translational Pain Research Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Sarah Roth
- MGH Translational Pain Research Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Marcos Vidal Melo
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Lucy Chen
- MGH Translational Pain Research Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Jianren Mao
- MGH Translational Pain Research Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| | - Jacob Hooker
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129
| | - Marco L Loggia
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129
| | - Yi Zhang
- MGH Translational Pain Research Center, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114
| |
Collapse
|
57
|
Wodarski R, Bagdas D, Paris JJ, Pheby T, Toma W, Xu R, Damaj MI, Knapp PE, Rice AS, Hauser KF. Reduced intraepidermal nerve fibre density, glial activation, and sensory changes in HIV type-1 Tat-expressing female mice: involvement of Tat during early stages of HIV-associated painful sensory neuropathy. Pain Rep 2018; 3:e654. [PMID: 29922746 PMCID: PMC5999412 DOI: 10.1097/pr9.0000000000000654] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/19/2018] [Accepted: 03/17/2018] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION HIV infection is associated with chronic pain states, including sensory neuropathy, which affects greater than 40% of patients. OBJECTIVES AND METHODS To determine the impact of HIV-Tat induction on nociceptive behaviour in female mice conditionally expressing HIV Tat1-86 protein through a doxycycline (DOX)-driven glial fibrillary acidic protein promoter, intraepidermal nerve fibre density and immune cell activation in the dorsal root ganglion (DRG) and spinal cord were assessed by immunohistochemistry. Mice were assessed for mechanical and thermal sensitivity for 9 weeks using von-Frey and Hargreaves tests. RESULTS Intraepidermal nerve fibre density was significantly reduced after 6 weeks of Tat induction, similar to sensory neuropathy seen in clinical HIV infection. Tat induction through DOX caused a significant reduction in paw withdrawal thresholds in a time-dependent manner starting the 4th week after Tat induction. No changes in paw withdrawal latencies were seen in Tat(-) control mice lacking the tat transgene. Although reductions in paw withdrawal thresholds increased throughout the study, no significant change in spontaneous motor activity was observed. Spinal cord (cervical and lumbar), DRG, and hind paw skin were collected at 8 days and 6 weeks after Tat induction. HIV-Tat mRNA expression was significantly increased in lumbar DRG and skin samples 8 days after DOX treatment. Tat induced a significant increase in the number of Iba-1 positive cells at 6 weeks, but not after 8 days, of exposure. No differences in glial fibrillary acidic protein immunoreactivity were observed. CONCLUSION These results suggest that Tat protein contributes to painful HIV-related sensory neuropathy during the initial stages of the pathogenesis.
Collapse
Affiliation(s)
- Rachel Wodarski
- Pain Research Group, Department of Surgery and Cancer, Imperial College, Chelsea and Westminster Hospital Campus, London, United Kingdom
| | - Deniz Bagdas
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason J. Paris
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of BioMolecular Sciences, University of Mississippi, University, MS, USA
| | - Tim Pheby
- Pain Research Group, Department of Surgery and Cancer, Imperial College, Chelsea and Westminster Hospital Campus, London, United Kingdom
| | - Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Ruqiang Xu
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew S.C. Rice
- Pain Research Group, Department of Surgery and Cancer, Imperial College, Chelsea and Westminster Hospital Campus, London, United Kingdom
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
58
|
Irvine KA, Sahbaie P, Liang DY, Clark JD. Traumatic Brain Injury Disrupts Pain Signaling in the Brainstem and Spinal Cord. J Neurotrauma 2018; 35:1495-1509. [PMID: 29373948 DOI: 10.1089/neu.2017.5411] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic pain is a common consequence of traumatic brain injury (TBI) that can increase the suffering of a patient and pose a significant challenge to rehabilitative efforts. Unfortunately, the mechanisms linking TBI to pain are poorly understood, and specific treatments for TBI-related pain are still lacking. Our laboratory has shown that TBI causes pain sensitization in areas distant to the site of primary injury, and that changes in spinal gene expression may underlie this sensitization. The aim of this study was to examine the roles that pain modulatory pathways descending from the brainstem play in pain after TBI. Deficiencies in one type of descending inhibition, diffuse noxious inhibitory control (DNIC), have been suggested to be responsible for the development of chronic pain by allowing excess and uncontrolled afferent nociceptive inputs. Here we expand our knowledge of pain after TBI in two ways: (1) by outlining the neuropathology in pain-related centers of the brain and spinal cord involved in DNIC using the rat lateral fluid percussion (LFP) model of TBI, and (2) by evaluating the effects of a potent histone acetyl transferase inhibitor, anacardic acid (AA), on LFP-induced pain behaviors and neuropathology when administered for several days after TBI. The results revealed that TBI induces transient mechanical allodynia and a chronic persistent loss of DNIC. Further, while short-term AA treatment can block acute nociceptive sensitization and some early neuropathological changes, this treatment neither prevented the loss of DNIC nor did it alter long-term neuropathological changes in the brain or spinal cord.
Collapse
Affiliation(s)
- Karen-Amanda Irvine
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| | - Peyman Sahbaie
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| | - De-Yong Liang
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| | - J David Clark
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| |
Collapse
|
59
|
Botulinum Toxin Type A-A Modulator of Spinal Neuron-Glia Interactions under Neuropathic Pain Conditions. Toxins (Basel) 2018; 10:toxins10040145. [PMID: 29614835 PMCID: PMC5923311 DOI: 10.3390/toxins10040145] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 12/29/2022] Open
Abstract
Neuropathic pain represents a significant clinical problem because it is a chronic condition often refractory to available therapy. Therefore, there is still a strong need for new analgesics. Botulinum neurotoxin A (BoNT/A) is used to treat a variety of clinical diseases associated with pain. Glia are in continuous bi-directional communication with neurons to direct the formation and refinement of synaptic connectivity. This review addresses the effects of BoNT/A on the relationship between glia and neurons under neuropathic pain. The inhibitory action of BoNT/A on synaptic vesicle fusion that blocks the release of miscellaneous pain-related neurotransmitters is known. However, increasing evidence suggests that the analgesic effect of BoNT/A is mediated through neurons and glial cells, especially microglia. In vitro studies provide evidence that BoNT/A exerts its anti-inflammatory effect by diminishing NF-κB, p38 and ERK1/2 phosphorylation in microglia and directly interacts with Toll-like receptor 2 (TLR2). Furthermore, BoNT/A appears to have no more than a slight effect on astroglia. The full activation of TLR2 in astroglia appears to require the presence of functional TLR4 in microglia, emphasizing the significant interaction between those cell types. In this review, we discuss whether and how BoNT/A affects the spinal neuron–glia interaction and reduces the development of neuropathy.
Collapse
|
60
|
Eitner A, Hofmann GO, Schaible HG. Mechanisms of Osteoarthritic Pain. Studies in Humans and Experimental Models. Front Mol Neurosci 2017; 10:349. [PMID: 29163027 PMCID: PMC5675866 DOI: 10.3389/fnmol.2017.00349] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022] Open
Abstract
Pain due to osteoarthritis (OA) is one of the most frequent causes of chronic pain. However, the mechanisms of OA pain are poorly understood. This review addresses the mechanisms which are thought to be involved in OA pain, derived from studies on pain mechanisms in humans and in experimental models of OA. Three areas will be considered, namely local processes in the joint associated with OA pain, neuronal mechanisms involved in OA pain, and general factors which influence OA pain. Except the cartilage all structures of the joints are innervated by nociceptors. Although the hallmark of OA is the degradation of the cartilage, OA joints show multiple structural alterations of cartilage, bone and synovial tissue. In particular synovitis and bone marrow lesions have been proposed to determine OA pain whereas the contribution of the other pathologies to pain generation has been studied less. Concerning the peripheral neuronal mechanisms of OA pain, peripheral nociceptive sensitization was shown, and neuropathic mechanisms may be involved at some stages. Structural changes of joint innervation such as local loss and/or sprouting of nerve fibers were shown. In addition, central sensitization, reduction of descending inhibition, descending excitation and cortical atrophies were observed in OA. The combination of different neuronal mechanisms may define the particular pain phenotype in an OA patient. Among mediators involved in OA pain, nerve growth factor (NGF) is in the focus because antibodies against NGF significantly reduce OA pain. Several studies show that neutralization of interleukin-1β and TNF may reduce OA pain. Many patients with OA exhibit comorbidities such as obesity, low grade systemic inflammation and diabetes mellitus. These comorbidities can significantly influence the course of OA, and pain research just began to study the significance of such factors in pain generation. In addition, psychologic and socioeconomic factors may aggravate OA pain, and in some cases genetic factors influencing OA pain were found. Considering the local factors in the joint, the neuronal processes and the comorbidities, a better definition of OA pain phenotypes may become possible. Studies are under way in order to improve OA and OA pain monitoring.
Collapse
Affiliation(s)
- Annett Eitner
- Department of Physiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Gunther O Hofmann
- Department of Traumatology and Orthopedic Surgery, University Hospital Jena, Friedrich Schiller University, Jena, Germany.,Trauma Center Bergmannstrost Halle, Halle, Germany
| | - Hans-Georg Schaible
- Department of Physiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
61
|
Microglia and astrocyte activation in the spinal cord of lame horses. Vet Anaesth Analg 2017; 45:92-102. [PMID: 29223561 DOI: 10.1016/j.vaa.2017.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine the microglial and astrocyte response to painful lameness in horses. STUDY DESIGN Ionized calcium binding adaptor molecule 1 (Iba-1) and glial fibrillary acidic protein (GFAP) expression, cell density and morphology were determined through immunofluorescence within the dorsal horn of equine spinal cord. ANIMALS A total of five adult horses with acute or chronic unilateral lameness, previously scheduled for euthanasia. METHODS Musculoskeletal lameness was evaluated in five horses through visual evaluation according to clinical guidelines. Spinal cord samples were obtained immediately after euthanasia, and distal limb lesions were confirmed through dissection and radiography. Iba-1 immunostaining was used for detection and characterization of dorsal horn microglia. GFAP was used for immunostaining of dorsal horn astrocytes. Iba-1 and GFAP labeled cells were quantified in the dorsal horn, and intensity of fluorescence was compared between the ipsi- and contralateral dorsal horn to the affected limb, and between dorsal horn segments of all horses. RESULTS Iba-1 expression was higher in the ipsilateral dorsal horn of the affected limb in contrast to the contralateral side dorsal horn. GFAP markers did not demonstrate increased astrocytic activity on the dorsal horn ipsilateral side to the distal limb lesion of affected horses. Horses with acute lameness predominantly had a spherical shape microglial phenotype, while cells from chronic lameness cases had variable morphology. Astrocytes evidenced small somas and large processes in both acute and chronic lameness, with higher GFAP localization in the main branches. As in the case of rodents, the localization of microglia and astrocytes in horses was mainly situated within laminae I, II and III. CONCLUSIONS AND CLINICAL RELEVANCE Iba-1 and GFAP are functional and morphological markers of spinal microglial cells and astrocytes in horses with lameness.
Collapse
|
62
|
Liver X Receptor α Is Involved in Counteracting Mechanical Allodynia by Inhibiting Neuroinflammation in the Spinal Dorsal Horn. Anesthesiology 2017; 127:534-547. [PMID: 28617705 DOI: 10.1097/aln.0000000000001718] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Liver X receptors, including α and β isoforms, are ligand-activated transcription factors. Whether liver X receptor α plays a role in neuropathic pain is unknown. METHODS A spared nerve injury model was established in adult male rats and mice. Von Frey tests were performed to evaluate the neuropathic pain behavior; Western blot and immunohistochemistry were performed to understand the underlying mechanisms. RESULTS Intrathecal injection of a specific liver X receptor agonist T0901317 or GW3965 could either prevent the development of mechanical allodynia or alleviate the established mechanical allodynia, both in rats and wild-type mice. GW3965 could inhibit the activation of glial cells and the expression of tumor necrosis factor-α (mean ± SD: 196 ± 48 vs. 119 ± 57; n = 6; P < 0.01) and interleukin 1β (mean ± SD: 215 ± 69 vs. 158 ± 74; n = 6; P < 0.01) and increase the expression of interleukin 10 in the spinal dorsal horn. All of the above effects of GW3965 could be abolished by liver X receptor α mutation. Moreover, more glial cells were activated, and more interleukin 1β was released in the spinal dorsal horn in liver X receptor α knockout mice than in wild-type mice after spared nerve injury. Aminoglutethimide, a neurosteroid synthesis inhibitor, blocked the inhibitory effect of T0901317 on mechanical allodynia, on the activation of glial cells, and on the expression of cytokines. CONCLUSIONS Activation of liver X receptor α inhibits mechanical allodynia by inhibiting the activation of glial cells and rebalancing cytokines in the spinal dorsal horn via neurosteroids.
Collapse
|
63
|
The Effect of Glucocorticoid and Glucocorticoid Receptor Interactions on Brain, Spinal Cord, and Glial Cell Plasticity. Neural Plast 2017; 2017:8640970. [PMID: 28928988 PMCID: PMC5591892 DOI: 10.1155/2017/8640970] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/14/2017] [Indexed: 12/15/2022] Open
Abstract
Stress, injury, and disease trigger glucocorticoid (GC) elevation. Elevated GCs bind to the ubiquitously expressed glucocorticoid receptor (GR). While GRs are in every cell in the nervous system, the expression level varies, suggesting that diverse cell types react differently to GR activation. Stress/GCs induce structural plasticity in neurons, Schwann cells, microglia, oligodendrocytes, and astrocytes as well as affect neurotransmission by changing the release and reuptake of glutamate. While general nervous system plasticity is essential for adaptation and learning and memory, stress-induced plasticity is often maladaptive and contributes to neuropsychiatric disorders and neuropathic pain. In this brief review, we describe the evidence that stress/GCs activate GR to promote cell type-specific changes in cellular plasticity throughout the nervous system.
Collapse
|
64
|
Haefeli J, Huie JR, Morioka K, Ferguson AR. Assessments of sensory plasticity after spinal cord injury across species. Neurosci Lett 2017; 652:74-81. [PMID: 28007646 PMCID: PMC5466896 DOI: 10.1016/j.neulet.2016.12.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/08/2016] [Accepted: 12/14/2016] [Indexed: 12/26/2022]
Abstract
Spinal cord injury (SCI) is a multifaceted phenomenon associated with alterations in both motor function and sensory function. A majority of patients with SCI report sensory disturbances, including not only loss of sensation, but in many cases enhanced abnormal sensation, dysesthesia and pain. Development of therapeutics to treat these abnormal sensory changes require common measurement tools that can enable cross-species translation from animal models to human patients. We review the current literature on translational nociception/pain measurement in SCI and discuss areas for further development. Although a number of tools exist for measuring both segmental and affective sensory changes, we conclude that there is a pressing need for better, integrative measurement of nociception/pain outcomes across species to enhance precise therapeutic innovation for sensory dysfunction in SCI.
Collapse
Affiliation(s)
- Jenny Haefeli
- Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| | - J Russell Huie
- Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| | - Kazuhito Morioka
- Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| | - Adam R Ferguson
- Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, USA; San Francisco Veteran's Affairs Medical Center, San Francisco, CA, USA.
| |
Collapse
|
65
|
Zhang J, Mense S, Treede RD, Hoheisel U. Prevention and reversal of latent sensitization of dorsal horn neurons by glial blockers in a model of low back pain in male rats. J Neurophysiol 2017; 118:2059-2069. [PMID: 28615336 DOI: 10.1152/jn.00680.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022] Open
Abstract
In an animal model of nonspecific low back pain, recordings from dorsal horn neurons were made to investigate the influence of glial cells in the central sensitization process. To induce a latent sensitization of the neurons, nerve growth factor (NGF) was injected into the multifidus muscle; the manifest sensitization to a second NGF injection 5 days later was used as a read-out. The sensitization manifested in increased resting activity and in an increased proportion of neurons responding to stimulation of deep somatic tissues. To block microglial activation, minocycline was continuously administered intrathecally starting 1 day before or 2 days after the first NGF injection. The glia inhibitor fluorocitrate that also blocks astrocyte activation was administrated 2 days after the first injection. Minocycline applied before the first NGF injection reduced the manifest sensitization after the second NGF injection to control values. The proportion of neurons responsive to stimulation of deep tissues was reduced from 50% to 17.7% (P < 0.01). No significant changes occurred when minocycline was applied after the first injection. In contrast, fluorocitrate administrated after the first NGF injection reduced significantly the proportion of neurons with deep input (15.8%, P < 0.01). A block of glia activation had no significant effect on the increased resting activity. The data suggest that blocking microglial activation prevented the NGF-induced latent spinal sensitization, whereas blocking astrocyte activation reversed it. The induction of spinal neuronal sensitization in this pain model appears to depend on microglia activation, whereas its maintenance is regulated by activated astrocytes.NEW & NOTEWORTHY Activated microglia and astrocytes mediate the latent sensitization induced by nerve growth factor in dorsal horn neurons that receive input from deep tissues of the low back. These processes may contribute to nonspecific low back pain.
Collapse
Affiliation(s)
- Juanjuan Zhang
- Chair of Neurophysiology, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and.,Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Siegfried Mense
- Chair of Neurophysiology, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and
| | - Rolf-Detlef Treede
- Chair of Neurophysiology, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and
| | - Ulrich Hoheisel
- Chair of Neurophysiology, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and
| |
Collapse
|
66
|
|
67
|
Abstract
More than 20% of adults worldwide experience different types of chronic pain, which are frequently associated with several comorbidities and a decrease in quality of life. Several approved painkillers are available, but current analgesics are often hampered by insufficient efficacy and/or severe adverse effects. Consequently, novel strategies for safe, highly efficacious treatments are highly desirable, particularly for chronic pain. Epigenetic mechanisms such as DNA methylation, histone modifications and microRNAs (miRNAs) strongly affect the regulation of gene expression, potentially for long periods over years or even generations, and have been associated with pathophysiological pain. Several studies, mostly in animals, revealed that inhibitors of DNA methylation, activators and inhibitors of histone modification and modulators of miRNAs reverse a number of pathological changes in the pain epigenome, which are associated with altered expression of pain-relevant genes. This epigenetic modulation might then reduce the nociceptive response and provide novel therapeutic options for analgesic therapy of chronic pain states. However, a number of challenges, such as nonspecific effects and poor delivery to target cells and tissues, hinder the rapid development of such analgesics. In this Review, we critically summarize data on epigenetics and pain, focusing on challenges in clinical development as well as possible new approaches to the drug modulation of the pain epigenome.
Collapse
Affiliation(s)
- Ellen Niederberger
- Pharmazentrum Frankfurt, Zentrum für Arzneimittelforschung Entwicklung und Sicherheit (ZAFES), Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | - Eduard Resch
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Project Group for Translational Medicine &Pharmacology, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Project Group for Translational Medicine &Pharmacology, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Pharmazentrum Frankfurt, Zentrum für Arzneimittelforschung Entwicklung und Sicherheit (ZAFES), Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology, Project Group for Translational Medicine &Pharmacology, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
68
|
Tran PB, Miller RE, Ishihara S, Miller RJ, Malfait AM. Spinal microglial activation in a murine surgical model of knee osteoarthritis. Osteoarthritis Cartilage 2017; 25:718-726. [PMID: 27646532 PMCID: PMC5354992 DOI: 10.1016/j.joca.2016.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 08/29/2016] [Accepted: 09/08/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Microgliosis, the activation of microglial cells, is thought to contribute to synaptic transmission in the dorsal horn and thereby promote chronic pain. The primary aim of this study was to document the temporal profile of dorsal horn microgliosis after destabilization of the medial meniscus (DMM) in wild type (WT) and Adamts5 null mice. Since neuronal fractalkine (CX3CL1) contributes to microgliosis, we assessed its release from dorsal root ganglia (DRG) cultures after DMM. DESIGN DMM or sham surgery was performed in the right knee of 10-week old male WT, CX3CR1-green fluorescent protein (GFP), or Adamts5 null C57BL/6 mice. Hind paw mechanical allodynia was monitored using von Frey fibers. L4 dorsal horn microgliosis was assessed 4, 8 and 16 weeks after surgery, based on the morphology of Iba1-immunoreactive microglia. DRG cells (L3-L5) were cultured and supernatants collected for fractalkine (FKN) ELISA. RESULTS In WT mice, numbers of activated microglia were increased 8 and 16 weeks, but not 4 weeks, after DMM but not sham surgery. DRG cultures showed increased basal FKN release at 8 and 16 weeks. Adamts5 null mice did not develop mechanical allodynia up to 16 weeks after DMM. Accordingly, DRG cultures from these mice did not exhibit increased FKN release and dorsal horn microgliosis did not occur. CONCLUSION DMM surgery leads to late stage dorsal horn microgliosis. The temporal correlation with DRG FKN release suggests it may contribute to microgliosis. Reduced microgliosis in Adamts5 null mice, which are protected from joint damage and associated mechanical allodynia after DMM, suggests that microgliosis is associated with joint damage and accompanying persistent pain.
Collapse
Affiliation(s)
- Phuong B. Tran
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL,Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL
| | | | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL,Department of Biochemistry, Rush University Medical Center, 1611 W. Harrison St, Suite 510, Chicago, IL,Address correspondence to: Anne-Marie Malfait, MD, PhD, Rush University Medical Center, 1611 W Harrison Street, suite 510, 60612 Chicago IL
| |
Collapse
|
69
|
Zhao H, Alam A, Chen Q, Eusman M, Pal A, Eguchi S, Wu L, Ma D. The role of microglia in the pathobiology of neuropathic pain development: what do we know? Br J Anaesth 2017; 118:504-516. [DOI: 10.1093/bja/aex006] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
70
|
Dauth S, Maoz BM, Sheehy SP, Hemphill MA, Murty T, Macedonia MK, Greer AM, Budnik B, Parker KK. Neurons derived from different brain regions are inherently different in vitro: a novel multiregional brain-on-a-chip. J Neurophysiol 2017; 117:1320-1341. [PMID: 28031399 PMCID: PMC5350271 DOI: 10.1152/jn.00575.2016] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/28/2016] [Accepted: 12/28/2016] [Indexed: 12/30/2022] Open
Abstract
Brain in vitro models are critically important to developing our understanding of basic nervous system cellular physiology, potential neurotoxic effects of chemicals, and specific cellular mechanisms of many disease states. In this study, we sought to address key shortcomings of current brain in vitro models: the scarcity of comparative data for cells originating from distinct brain regions and the lack of multiregional brain in vitro models. We demonstrated that rat neurons from different brain regions exhibit unique profiles regarding their cell composition, protein expression, metabolism, and electrical activity in vitro. In vivo, the brain is unique in its structural and functional organization, and the interactions and communication between different brain areas are essential components of proper brain function. This fact and the observation that neurons from different areas of the brain exhibit unique behaviors in vitro underline the importance of establishing multiregional brain in vitro models. Therefore, we here developed a multiregional brain-on-a-chip and observed a reduction of overall firing activity, as well as altered amounts of astrocytes and specific neuronal cell types compared with separately cultured neurons. Furthermore, this multiregional model was used to study the effects of phencyclidine, a drug known to induce schizophrenia-like symptoms in vivo, on individual brain areas separately while monitoring downstream effects on interconnected regions. Overall, this work provides a comparison of cells from different brain regions in vitro and introduces a multiregional brain-on-a-chip that enables the development of unique disease models incorporating essential in vivo features.NEW & NOTEWORTHY Due to the scarcity of comparative data for cells from different brain regions in vitro, we demonstrated that neurons isolated from distinct brain areas exhibit unique behaviors in vitro. Moreover, in vivo proper brain function is dependent on the connection and communication of several brain regions, underlining the importance of developing multiregional brain in vitro models. We introduced a novel brain-on-a-chip model, implementing essential in vivo features, such as different brain areas and their functional connections.
Collapse
Affiliation(s)
- Stephanie Dauth
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Ben M Maoz
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Sean P Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Matthew A Hemphill
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Tara Murty
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Mary Kate Macedonia
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Angie M Greer
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| | - Bogdan Budnik
- Mass Spectrometry and Proteomics Resource Laboratory, Harvard University, Cambridge, Massachusetts
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts; and
| |
Collapse
|
71
|
Lu C, Shi L, Sun B, Zhang Y, Hou B, Sun Y, Ma Z, Gu X. A Single Intrathecal or Intraperitoneal Injection of CB2 Receptor Agonist Attenuates Bone Cancer Pain and Induces a Time-Dependent Modification of GRK2. Cell Mol Neurobiol 2017; 37:101-109. [PMID: 26935064 PMCID: PMC11482228 DOI: 10.1007/s10571-016-0349-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 02/11/2016] [Indexed: 01/01/2023]
Abstract
The objective of this study was to explore the potential role of G-protein-coupled receptor kinase 2 (GRK2) in the progression of cannabinoid 2 receptor (CB2) agonist-induced analgesic effects of bone cancer pain. Female Sprague-Dawley rats, weighing 160-180 g, were utilized to establish a model of bone cancer pain induced by intra-tibia inoculation of Walker 256 mammary gland carcinoma cells. JWH-015, a selective CB2 agonist, was injected intrathecally or intraperitoneally on postoperative day 10. Bone cancer-induced pain behaviors-mechanical allodynia and ambulatory pain-were assessed on postoperative days -1 (baseline), 4, 7, and 10 and at post-treatment hours 2, 6, 24, 48, and 72. The expressions of spinal CB2 and GRK2 protein were detected by Western Blotting on postoperative days -1 (baseline), 4, 7, and 10 and at post-treatment hours 6, 24, and 72. The procedure produced prolonged mechanical allodynia, ambulatory pain, and different changes in spinal CB2 and GRK2 expression levels. Intrathecal or intraperitoneal administration of JWH-015 alleviated the induced mechanical allodynia and ambulatory pain, and inhibited the downregulation of spinal GRK2 expression. These effects were in a time-dependent manner and reversed by pretreatment of CB2 selective antagonist AM630. The results affirmed CB2 receptor agonists might serve as new treatment targets for bone cancer pain. Moreover, spinal GRK2 was an important regulator of CB2 receptor agonist-analgesia pathway.
Collapse
Affiliation(s)
- Cui'e Lu
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Linyu Shi
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Bei Sun
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Yu Zhang
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Bailing Hou
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Yu'e Sun
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum-Tower Hospital of Medical College of Nanjing University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
72
|
Tateda S, Kanno H, Ozawa H, Sekiguchi A, Yahata K, Yamaya S, Itoi E. Rapamycin suppresses microglial activation and reduces the development of neuropathic pain after spinal cord injury. J Orthop Res 2017; 35:93-103. [PMID: 27279283 DOI: 10.1002/jor.23328] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 06/05/2016] [Indexed: 02/04/2023]
Abstract
Rapamycin is an inhibitor of the mammalian target of rapamycin (mTOR) signaling pathway, plays an important role in multiple cellular functions. Our previous study showed rapamycin treatment in acute phase reduced the neural tissue damage and locomotor impairment after spinal cord injury (SCI). However, there has been no study to investigate the therapeutic effect of rapamycin on neuropathic pain after SCI. In this study, we examined whether rapamycin reduces neuropathic pain following SCI in mice. We used a mouse model of thoracic spinal cord contusion injury, and divided the mice into the rapamycin-treated and the vehicle-treated groups. The rapamycin-treated mice were intraperitoneally injected with rapamycin (1 mg/kg) 4 h after SCI. The rapamycin treatment suppressed phosphorylated-p70S6K in the injured spinal cord that indicated inhibition of mTOR. The rapamycin treatment significantly improved not only locomotor function, but also mechanical and thermal hypersensitivity in the hindpaws after SCI. In an immunohistochemical analysis, Iba-1-stained microglia in the lumbar spinal cord was significantly decreased in the rapamycin-treated mice. In addition, the activity of p38 MAPK in the lumbar spinal cord was significantly attenuated by rapamycin treatment. Furthermore, phosphorylated-p38 MAPK-positive microglia was relatively decreased in the rapamycin-treated mice. These results indicated rapamycin administration in acute phase to reduce secondary neural tissue damage can contribute to the suppression of the microglial activation in the lumbar spinal cord and attenuate the development of neuropathic pain after SCI. The present study first demonstrated that rapamycin has significant therapeutic potential to reduce the development of neuropathic pain following SCI. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:93-103, 2017.
Collapse
Affiliation(s)
- Satoshi Tateda
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| | - Haruo Kanno
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| | - Hiroshi Ozawa
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| | - Akira Sekiguchi
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| | - Kenichiro Yahata
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| | - Seiji Yamaya
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Miyagi, Japan
| |
Collapse
|
73
|
NADPH-diaphorase reactivity and Fos-immunoreactivity within the ventral horn of the lumbar spinal cord of cats submitted to acute muscle inflammation induced by injection of carrageenan. Acta Histochem 2016; 118:659-664. [PMID: 27692234 DOI: 10.1016/j.acthis.2016.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/14/2016] [Accepted: 09/19/2016] [Indexed: 11/21/2022]
Abstract
The NADPH-diaphorase activity and Fos-immunoreactivity within the ventral horn of the lumbar spinal cord were studied in cats with acute unilateral myositis following injection of carrageenan into the m.m. gastrocnemius-soleus. In carrageenan-injected cats maximum in the mean number of intensely stained NADPH-diaphorase reactive (NADPH-dr) neurons was found in lamina VII (+100%) and VIII (+33%) of the contralateral ventral horn of the L6/L7 segments as compared with control animals. The maximumal level of Fos-immunoreactivity was registered in the same laminae with ipsilateral predominance (39.3±4.6 and 7.6±0.9 cells), in comparison with the contralateral side (13.6±0.8 and 5.5±0.6 cells, respectively; P<0.05). We also visualized low-intensely stained and double labelled (Fos immunoreactive+low-intensely stained NADPH-dr) multipolar and fusiform Renshaw-like cells (RLCs) within the ventral horn on both sides of the L6/L7 segments in carrageenan-injected cats. We visualized the double labelled (Fos-ir+NADPH-dr) multipolar and fusiform Renshaw-like cells (RLCs) within the ventral horn on both sides of the L6/L7 segments in carrageenan-injected cats. A significant difference in the mean number of RLCs was recorded between the ipsi- and contralateral sides in the lamina VII (13.6±2.5 vs. 4.9±0.7 cells, respectively). We suppose that activation of inhibitory RLCs in ipsilateral lamina VII could be directed on attenuation of activation of motoneurons during muscle pain development. Our study showed that a significant contralateral increase in the number of NADPH-dr cells is accompanied by an ipsilateral increase in c-Fos expression in lamina VII. These data may suggest that NADPH-dr neurons of the contralateral ventral horn through commissural connections also involved in the maintenance of the neuronal activity associated with acute muscle inflammation. It is also hypothesized, that during acute myositis, plastic changes in the ventral horn activate the processes of disinhibition due to an increase in the number of NADPH-d-reactive neurons in the spinal gray matter.
Collapse
|
74
|
Maraviroc reduces neuropathic pain through polarization of microglia and astroglia – Evidence from in vivo and in vitro studies. Neuropharmacology 2016; 108:207-19. [DOI: 10.1016/j.neuropharm.2016.04.024] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/13/2016] [Accepted: 04/19/2016] [Indexed: 02/08/2023]
|
75
|
Differential Changes in Neuronal Excitability in the Spinal Dorsal Horn After Spinal Nerve Ligation in Rats. Neurochem Res 2016; 41:2880-2889. [DOI: 10.1007/s11064-016-2003-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/05/2016] [Accepted: 07/07/2016] [Indexed: 12/14/2022]
|
76
|
Xu D, Omura T, Masaki N, Arima H, Banno T, Okamoto A, Hanada M, Takei S, Matsushita S, Sugiyama E, Setou M, Matsuyama Y. Increased arachidonic acid-containing phosphatidylcholine is associated with reactive microglia and astrocytes in the spinal cord after peripheral nerve injury. Sci Rep 2016; 6:26427. [PMID: 27210057 PMCID: PMC4876408 DOI: 10.1038/srep26427] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/28/2016] [Indexed: 12/30/2022] Open
Abstract
Peripheral nerve injury (PNI) triggers cellular and molecular changes in the spinal cord. However, little is known about how the polyunsaturated fatty acid-containing phosphatidylcholines (PUFA-PCs) are regulated in the spinal cord after PNI and the association of PUFA-PCs with the non-neuronal cells within in the central nervous system (CNS). In this study, we found that arachidonic acid-containing phosphatidylcholine (AA-PC), [PC(16:0/20:4)+K](+), was significantly increased in the ipsilateral ventral and dorsal horns of the spinal cord after sciatic nerve transection, and the increased expression of [PC(16:0/20:4)+K](+) spatiotemporally resembled the increase of reactive microglia and the astrocytes. From the lipidomics point of view, we conclude that [PC(16:0/20:4)+K](+) could be the main phospholipid in the spinal cord influenced by PNI, and the regulation of specific phospholipid molecule in the CNS after PNI is associated with the reactive microglia and astrocytes.
Collapse
Affiliation(s)
- Dongmin Xu
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takao Omura
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Noritaka Masaki
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hideyuki Arima
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tomohiro Banno
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Ayako Okamoto
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Mitsuru Hanada
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Shiro Takei
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Shoko Matsushita
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Eiji Sugiyama
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Mitsutoshi Setou
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong, 999077 China
- Division of Neural Systematics, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Yukihiro Matsuyama
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
77
|
Amitriptyline, minocycline and maropitant reduce the sevoflurane minimum alveolar concentration and potentiate remifentanil but do not prevent acute opioid tolerance and hyperalgesia in the rat: a randomised laboratory study. Eur J Anaesthesiol 2016; 32:248-54. [PMID: 24849503 DOI: 10.1097/eja.0000000000000098] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The antidepressant amitriptyline, the inhibitor of microglia activation minocycline, and the neurokinin-1 antagonist maropitant have all been used to prevent or treat hyperalgesia and opioid tolerance. OBJECTIVES To determine the effect of amitriptyline, minocycline, maropitant, independently or with remifentanil, on the sevoflurane minimum alveolar concentration in rats and whether these drugs may block opioid-induced hyperalgesia and acute opioid tolerance under inhalational anaesthesia. DESIGN A randomised, laboratory study. SETTING Experimental Unit, La Paz University Hospital, Madrid, Spain. ANIMALS One hundred and fourteen adult male Wistar rats. INTERVENTIONS Intraperitoneal administration of amitriptyline (10 and 50 mg kg-1), minocycline (30 and 100 mg kg-1), maropitant (10 and 30 mg kg-1) or isotonic saline, combined with a constant rate intravenous infusion of remifentanil (240 μg kg-1 h-1) or saline. MAIN OUTCOME MEASURES Sevoflurane minimum alveolar concentration was determined before and after administration of the drugs; acute opioid tolerance was defined as a decreased ability of remifentanil to reduce the minimum alveolar concentration in the short term. In addition, mechanical nociceptive thresholds were determined before and after these treatments. Opioid-induced hyperalgesia was defined as an increase in mechanical nociceptive thresholds after opioid administration. RESULTS Amitriptyline, minocycline and maropitant reduced minimum alveolar concentration up to 24 (8)%, 23 (6)% and 15 (5)%, respectively (P <0.001). Remifentanil alone reduced minimum alveolar concentration by 36 (6)% (P <0.001), and in combination with amitriptyline, minocycline and maropitant, the reduction was 76 (9)%, 75 (16)% and 59 (5)%, respectively (P <0.001). An acute tolerance effect (P < 0.01) and a decrease in the mechanical nociceptive thresholds were observed with remifentanil in all groups. CONCLUSION Amitriptyline, minocycline and maropitant reduced the minimum alveolar concentration and potentiated the remifentanil minimum alveolar concentration reduction but failed to block opioid-induced hyperalgesia and acute opioid tolerance.
Collapse
|
78
|
Kami K, Taguchi Ms S, Tajima F, Senba E. Improvements in impaired GABA and GAD65/67 production in the spinal dorsal horn contribute to exercise-induced hypoalgesia in a mouse model of neuropathic pain. Mol Pain 2016; 12:12/0/1744806916629059. [PMID: 27030712 PMCID: PMC4956002 DOI: 10.1177/1744806916629059] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/04/2015] [Indexed: 11/16/2022] Open
Abstract
Background Physical exercise effectively attenuates neuropathic pain, and multiple events including the inhibition of activated glial cells in the spinal dorsal horn, activation of the descending pain inhibitory system, and reductions in pro-inflammatory cytokines in injured peripheral nerves may contribute to exercise-induced hypoalgesia. Since fewer GABAergic hypoalgesic interneurons exist in the dorsal horn in neuropathic pain model animals, the recovery of impaired GABAergic inhibition in the dorsal horn may improve pain behavior. We herein determined whether the production of gamma-aminobutyric acid (GABA) and glutamic acid decarboxylase (GAD) in the dorsal horn is restored by treadmill running and contributes to exercise-induced hypoalgesia in neuropathic pain model mice. C57BL/6 J mice underwent partial sciatic nerve ligation (PSL). PSL-Runner mice ran on a treadmill at 7 m/min for 60 min/day, 5 days/week, from two days after PSL. Results Mechanical allodynia and heat hyperalgesia developed in PSL-Sedentary mice but were significantly attenuated in PSL-Runner mice. PSL markedly decreased GABA and GAD65/67 levels in neuropils in the ipsilateral dorsal horn, while treadmill running inhibited these reductions. GABA+ neuronal nuclei+ interneuron numbers in the ipsilateral dorsal horn were significantly decreased in PSL-Sedentary mice but not in PSL-Runner mice. Pain behavior thresholds positively correlated with GABA and GAD65/67 levels and GABAergic interneuron numbers in the ipsilateral dorsal horns of PSL-Sedentary and -Runner mice. Conclusions Treadmill running prevented PSL-induced reductions in GAD65/67 production, and, thus, GABA levels may be retained in interneurons and neuropils in the superficial dorsal horn. Therefore, improvements in impaired GABAergic inhibition may be involved in exercise-induced hypoalgesia.
Collapse
Affiliation(s)
- Katsuya Kami
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Satoru Taguchi Ms
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Fumihiro Tajima
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Emiko Senba
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama City, Wakayama, Japan Department of Physical Therapy, Osaka Yukioka College of Health Science, Ibaraki City, Osaka, Japan
| |
Collapse
|
79
|
Kami K, Taguchi S, Tajima F, Senba E. Histone Acetylation in Microglia Contributes to Exercise-Induced Hypoalgesia in Neuropathic Pain Model Mice. THE JOURNAL OF PAIN 2016; 17:588-99. [PMID: 26844418 DOI: 10.1016/j.jpain.2016.01.471] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 12/20/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED Physical exercise can attenuate neuropathic pain (NPP), but the exact mechanism underlying exercise-induced hypoalgesia (EIH) remains unclear. Recent studies have shown that histone hyperacetylation via pharmacological inhibition of histone deacetylases in the spinal cord attenuates NPP, and that histone acetylation may lead to the production of analgesic factors including interleukin 10. We intended to clarify whether histone acetylation in microglia in the spinal dorsal horn contributes to EIH in NPP model mice. C57BL/6J mice underwent partial sciatic nerve ligation (PSL) and PSL- and sham-runner mice ran on a treadmill at a speed of 7 m/min for 60 min/d, 5 days per week, from 2 days after the surgery. PSL-sedentary mice developed mechanical allodynia and heat hyperalgesia, but such behaviors were significantly attenuated in PSL-runner mice. In immunofluorescence analysis, PSL surgery markedly increased the number of histone deacetylase 1-positive/CD11b-positive microglia in the ipsilateral superficial dorsal horn, and they were significantly decreased by treadmill-running. Moreover, the number of microglia with nuclear expression of acetylated H3K9 in the ipsilateral superficial dorsal horn was maintained at low levels in PSL-sedentary mice, but running exercise significantly increased them. Therefore, we conclude that the epigenetic modification that causes hyperacetylation of H3K9 in activated microglia may play a role in producing EIH. PERSPECTIVE This article presents the importance of epigenetic modification in microglia in producing EIH. The current research is not only helpful for developing novel nonpharmacological therapy for NPP, but will also enhance our understanding of the mechanisms and availability of exercise in our daily life.
Collapse
Affiliation(s)
- Katsuya Kami
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Satoru Taguchi
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan
| | - Fumihiro Tajima
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan
| | - Emiko Senba
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan; Department of Physical Therapy, Osaka Yukioka College of Health Science, Osaka, Japan
| |
Collapse
|
80
|
Segond von Banchet G, König C, Patzer J, Eitner A, Leuchtweis J, Ebbinghaus M, Boettger MK, Schaible HG. Long-Lasting Activation of the Transcription Factor CREB in Sensory Neurons by Interleukin-1β During Antigen-Induced Arthritis in Rats: A Mechanism of Persistent Arthritis Pain? Arthritis Rheumatol 2016; 68:532-41. [PMID: 26473326 DOI: 10.1002/art.39445] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/17/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE In spite of successful treatment of immune-mediated arthritis, many patients still experience pain. We undertook this study to investigate whether antigen-induced arthritis (AIA) in rats triggers neuronal changes in sensory neurons that outlast the inflammatory process. METHODS We induced unilateral AIA in the knee joint and assessed in sensory neurons the expression of CREB, a transcription factor that regulates genes involved in neuronal plasticity. We tested whether neutralization of the effects of tumor necrosis factor (TNF) by etanercept or infliximab or neutralization of the effects of interleukin-1β (IL-1β) by anakinra influences the up-regulation of phospho-CREB, and we studied the up-regulation of phospho-CREB by IL-1β and TNF in cultured dorsal root ganglion (DRG) neurons. RESULTS Unilateral AIA caused bilateral up-regulation of phospho-CREB in lumbar DRG neurons. While inflammation and pain subsided within 21 days, the up-regulation of phospho-CREB still persisted on day 42. At this time point mechanical hyperalgesia at the knee reappeared in the absence of swelling. TNF neutralization during AIA significantly reduced pain-related behavior but did not prevent phospho-CREB up-regulation. In contrast, anakinra, which only reduced thermal hyperalgesia, prevented phospho-CREB up-regulation, suggesting a role of IL-1β in this process. In cultured DRG neurons the application of IL-1β significantly enhanced phospho-CREB. CONCLUSION Immune-mediated arthritis causes neuroplastic changes in sensory neurons that outlast the inflammatory phase. Such changes may facilitate the persistence or recurrence of pain after remission of arthritis. IL-1β is an important trigger in this process, although its neutralization barely reduced mechanical hyperalgesia during inflammation.
Collapse
Affiliation(s)
| | - Christian König
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | - Jessica Patzer
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | - Annett Eitner
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | - Johannes Leuchtweis
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | - Matthias Ebbinghaus
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | - Michael K Boettger
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | - Hans-Georg Schaible
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| |
Collapse
|
81
|
Popiolek-Barczyk K, Mika J. Targeting the Microglial Signaling Pathways: New Insights in the Modulation of Neuropathic Pain. Curr Med Chem 2016; 23:2908-2928. [PMID: 27281131 PMCID: PMC5427777 DOI: 10.2174/0929867323666160607120124] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/23/2016] [Accepted: 06/06/2016] [Indexed: 12/30/2022]
Abstract
The microglia, once thought only to be supporting cells of the central nervous system (CNS), are now recognized to play essential roles in many pathologies. Many studies within the last decades indicated that the neuro-immune interaction underlies the generation and maintenance of neuropathic pain. Through a large number of receptors and signaling pathways, the microglial cells communicate with neurons, astrocytes and other cells, including those of the immune system. A disturbance or loss of CNS homeostasis causes rapid responses of the microglia, which undergo a multistage activation process. The activated microglia change their cell shapes and gene expression profiles, which induce proliferation, migration, and the production of pro- or antinociceptive factors. The cells release a large number of mediators that can act in a manner detrimental or beneficial to the surrounding cells and can indirectly alter the nociceptive signals. This review discusses the most important microglial intracellular signaling cascades (MAPKs, NF-kB, JAK/STAT, PI3K/Akt) that are essential for neuropathic pain development and maintenance. Our objective was to identify new molecular targets that may result in the development of powerful tools to control the signaling associated with neuropathic pain.
Collapse
Affiliation(s)
| | - Joanna Mika
- Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str., 31-343 Krakow, Poland.
| |
Collapse
|
82
|
Central nervous system myeloid cells as drug targets: current status and translational challenges. Nat Rev Drug Discov 2015; 15:110-24. [DOI: 10.1038/nrd.2015.14] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
83
|
Abstract
G-protein receptor 84 (GPR84) is an orphan receptor that is induced markedly in monocytes/macrophages and microglia during inflammation, but its pathophysiological function is unknown. Here, we investigate the role of GPR84 in a murine model of traumatic nerve injury. Naive GPR84 knock-out (KO) mice exhibited normal behavioral responses to acute noxious stimuli, but subsequent to partial sciatic nerve ligation (PNL), KOs did not develop mechanical or thermal hypersensitivity, in contrast to wild-type (WT) littermates. Nerve injury increased ionized calcium binding adapter molecule 1 (Iba1) and phosphorylated p38 MAPK immunoreactivity in the dorsal horn and Iba1 and cluster of differentiation 45 expression in the sciatic nerve, with no difference between genotypes. PCR array analysis revealed that Gpr84 expression was upregulated in the spinal cord and sciatic nerve of WT mice. In addition, the expression of arginase-1, a marker for anti-inflammatory macrophages, was upregulated in KO sciatic nerve. Based on this evidence, we investigated whether peripheral macrophages behave differently in the absence of GPR84. We found that lipopolysaccharide-stimulated KO macrophages exhibited attenuated expression of several proinflammatory mediators, including IL-1β, IL-6, and TNF-α. Forskolin-stimulated KO macrophages also showed greater cAMP induction, a second messenger associated with immunosuppression. In summary, our results demonstrate that GPR84 is a proinflammatory receptor that contributes to nociceptive signaling via the modulation of macrophages, whereas in its absence the response of these cells to an inflammatory insult is impaired.
Collapse
|
84
|
Abstract
Proinflammatory cytokines are major mediators in the pathogenesis of diseases of joints such as rheumatoid arthritis and osteoarthritis. This review emphasizes that proinflammatory cytokines such as tumor necrosis factor-alpha, interleukin-1beta, interleukin-6 and interleukin-17 are also mediators of pain by directly acting on the nociceptive system. Proportions of nociceptive sensory neurons express receptors for these cytokines, and the application of cytokines rapidly changes the excitability, ion currents and second messenger systems of these neurons. By inducing persistent sensitization of nociceptive sensory neurons (C- and a proportion of Aδ-fibers) for mechanical stimuli in the joint (a process called peripheral sensitization), these cytokines significantly contribute to the persistent hyperalgesia typical for many disease states of the joint. In addition, the disease-associated release of cytokines in the spinal cord supports the generation of central sensitization. The therapeutic neutralization of proinflammatory cytokines thus not only reduces the process of inflammation but may directly reduce hyperalgesia and pain by reversing the neuronal effects of cytokines. It is emerging that different cytokines have different actions on neurons. The neutralization of tumor necrosis factor-alpha reduces both mechanical and thermal hyperalgesia of the joint. The neutralization of interleukin-1beta attenuates thermal hyperalgesia whereas the neutralization of interleukin-6 and interleukin-17 mainly reduces mechanical hyperalgesia. These different effects are partly explained by influencing different target molecules in sensory neurons. For example, in cultured sensory neurons tumor necrosis factor-alpha and interleukin-1beta upregulate the TRPV1 ion channel, which is involved in the transduction of heat stimuli, consistent with an effect of these cytokines in thermal hyperalgesia. By contrast, interleukin-17 upregulates the TRPV4 ion channel, which has a role in the transduction of mechanical stimuli. Thus, the analgesic potential of neutralizing cytokines seems to depend on which cytokine is mainly involved in the particular pain state.
Collapse
Affiliation(s)
- Hans-Georg Schaible
- Institute of Physiology I/Neurophysiology, Jena University Hospital - Friedrich Schiller University, Teichgraben 8, Jena, D-07740, Germany.
| |
Collapse
|
85
|
Nazemi S, Manaheji H, Noorbakhsh SM, Zaringhalam J, Sadeghi M, Mohammad-Zadeh M, Haghparast A. Inhibition of microglial activity alters spinal wide dynamic range neuron discharge and reduces microglial Toll-like receptor 4 expression in neuropathic rats. Clin Exp Pharmacol Physiol 2015; 42:772-9. [DOI: 10.1111/1440-1681.12414] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/15/2015] [Accepted: 04/18/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Samad Nazemi
- Department of Physiology; Sabzevar University of Medical Sciences; Sabzevar Iran
- Department of Neurophysiology; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Homa Manaheji
- Department of Neurophysiology; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | | | - Jalal Zaringhalam
- Department of Neurophysiology; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mehdi Sadeghi
- Department of Physiology; Bushehr University of Medical Sciences; Bushehr Iran
| | | | - Abbas Haghparast
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
86
|
Ogbonna AC, Clark AK, Malcangio M. Development of monosodium acetate-induced osteoarthritis and inflammatory pain in ageing mice. AGE (DORDRECHT, NETHERLANDS) 2015; 37:9792. [PMID: 25971876 PMCID: PMC4430498 DOI: 10.1007/s11357-015-9792-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 05/04/2015] [Indexed: 05/05/2023]
Abstract
Most conditions associated with ageing result from an age-related loss in the function of cells and tissues that maintain body homeostasis. In osteoarthritis (OA) patients, an inadequate response to stress or joint injury can lead to tissue destruction which can result in chronic pain. Here, we evaluated the development of monoiodoacetate (MIA)-induced OA in 3-, 15- and 22-month-old mice and assessed the pain-like behaviours and the spinal microglial changes associated with MIA administration. We observed that in aged mice, nocifensive behaviour was significantly attenuated in comparison to young adults despite similar knee joint pathology. Specifically referred mechanical allodynia associated with the MIA initial inflammatory phase (0-10 days) was significantly attenuated in 22-month-old mice. In contrast, the late phase of MIA-induced mechanical allodynia was comparable between age groups. Significant increase of microglia cell numbers was detected in 3, but not 15- and 22-month-old spinal cords. Furthermore, in the zymosan model of acute inflammation, mechanical allodynia was attenuated, and microglial response was less robust in 22 compared to 3-month-old mice. This study suggests that nocifensive responses to damaging stimuli are altered with advancing age and microglial response to peripheral damage is less robust.
Collapse
Affiliation(s)
- Andrea C. Ogbonna
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London Bridge, London, SE1 1UL UK
| | - Anna K. Clark
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London Bridge, London, SE1 1UL UK
| | - Marzia Malcangio
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London Bridge, London, SE1 1UL UK
| |
Collapse
|
87
|
Foster E, Wildner H, Tudeau L, Haueter S, Ralvenius WT, Jegen M, Johannssen H, Hösli L, Haenraets K, Ghanem A, Conzelmann KK, Bösl M, Zeilhofer HU. Targeted ablation, silencing, and activation establish glycinergic dorsal horn neurons as key components of a spinal gate for pain and itch. Neuron 2015; 85:1289-304. [PMID: 25789756 PMCID: PMC4372258 DOI: 10.1016/j.neuron.2015.02.028] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/26/2015] [Accepted: 02/12/2015] [Indexed: 01/28/2023]
Abstract
The gate control theory of pain proposes that inhibitory neurons of the spinal dorsal horn exert critical control over the relay of nociceptive signals to higher brain areas. Here we investigated how the glycinergic subpopulation of these neurons contributes to modality-specific pain and itch processing. We generated a GlyT2::Cre transgenic mouse line suitable for virus-mediated retrograde tracing studies and for spatially precise ablation, silencing, and activation of glycinergic neurons. We found that these neurons receive sensory input mainly from myelinated primary sensory neurons and that their local toxin-mediated ablation or silencing induces localized mechanical, heat, and cold hyperalgesia; spontaneous flinching behavior; and excessive licking and biting directed toward the corresponding skin territory. Conversely, local pharmacogenetic activation of the same neurons alleviated neuropathic hyperalgesia and chloroquine- and histamine-induced itch. These results establish glycinergic neurons of the spinal dorsal horn as key elements of an inhibitory pain and itch control circuit. Glycinergic dorsal horn neurons exert segmental control over pain and itch Their local inhibition causes hyperalgesia and signs of spontaneous discomfort Local activation reduces acute pain, neuropathic hyperalgesia, and chemical itch
Collapse
Affiliation(s)
- Edmund Foster
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Laetitia Tudeau
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir Prelog Weg 4, 8093 Zürich, Switzerland
| | - Sabine Haueter
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir Prelog Weg 4, 8093 Zürich, Switzerland
| | - William T Ralvenius
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Monika Jegen
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir Prelog Weg 4, 8093 Zürich, Switzerland
| | - Helge Johannssen
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Ladina Hösli
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Karen Haenraets
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir Prelog Weg 4, 8093 Zürich, Switzerland
| | - Alexander Ghanem
- Gene Center, Ludwig Maximilians University Munich, Max von Pettenkofer Institute of Virology, Feodor Lynen Strasse 25, 81377 Munich, Germany
| | - Karl-Klaus Conzelmann
- Gene Center, Ludwig Maximilians University Munich, Max von Pettenkofer Institute of Virology, Feodor Lynen Strasse 25, 81377 Munich, Germany
| | - Michael Bösl
- Experimental Biomedicine, University of Würzburg, Josef Schneider Strasse 2, 97080 Würzburg, Germany
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir Prelog Weg 4, 8093 Zürich, Switzerland.
| |
Collapse
|
88
|
Perera CJ, Duffy SS, Lees JG, Kim CF, Cameron B, Apostolopoulos V, Moalem-Taylor G. Active immunization with myelin-derived altered peptide ligand reduces mechanical pain hypersensitivity following peripheral nerve injury. J Neuroinflammation 2015; 12:28. [PMID: 25885812 PMCID: PMC4340611 DOI: 10.1186/s12974-015-0253-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/22/2015] [Indexed: 11/10/2022] Open
Abstract
Background T cells have been implicated in neuropathic pain that is caused by peripheral nerve injury. Immunogenic myelin basic protein (MBP) peptides have been shown to initiate mechanical allodynia in a T cell-dependent manner. Antagonistic altered peptide ligands (APLs) are peptides with substitutions in amino acid residues at T cell receptor contact sites and can inhibit T cell function and modulate inflammatory responses. In the present study, we studied the effects of immunization with MBP-derived APL on pain behavior and neuroinflammation in an animal model of peripheral nerve injury. Methods Lewis rats were immunized subcutaneously at the base of the tail with either a weakly encephalitogenic peptide of MBP (cyclo-MBP87-99) or APL (cyclo-(87-99)[A91,A96]MBP87-99) in complete Freund’s adjuvant (CFA) or CFA only (control), following chronic constriction injury (CCI) of the left sciatic nerve. Pain hypersensitivity was tested by measurements of paw withdrawal threshold to mechanical stimuli, regulatory T cells in spleen and lymph nodes were analyzed by flow cytometry, and immune cell infiltration into the nervous system was assessed by immunohistochemistry (days 10 and 30 post-CCI). Cytokines were measured in serum and nervous tissue of nerve-injured rats (day 10 post-CCI). Results Rats immunized with the APL cyclo-(87-99)[A91,A96]MBP87-99 had significantly reduced mechanical pain hypersensitivity in the ipsilateral hindpaw compared to cyclo-MBP87-99-treated and control rats. This was associated with significantly decreased infiltration of T cells and ED1+ macrophages in the injured nerve of APL-treated animals. The percentage of anti-inflammatory (M2) macrophages was significantly upregulated in the APL-treated rats on day 30 post-CCI. Compared to the control rats, microglial activation in the ipsilateral lumbar spinal cord was significantly increased in the MBP-treated rats, but was not altered in the rats immunized with the MBP-derived APL. In addition, immunization with the APL significantly increased splenic regulatory T cells. Several cytokines were significantly altered after CCI, but no significant difference was observed between the APL-treated and control rats. Conclusions These results suggest that immune deviation by active immunization with a non-encephalitogenic MBP-derived APL mediates an analgesic effect in animals with peripheral nerve injury. Thus, T cell immunomodulation warrants further investigation as a possible therapeutic strategy for the treatment of peripheral neuropathic pain.
Collapse
Affiliation(s)
- Chamini J Perera
- School of Medical Sciences, University of New South Wales, UNSW Medicine, Sydney, NSW, 2052, Australia.
| | - Samuel S Duffy
- School of Medical Sciences, University of New South Wales, UNSW Medicine, Sydney, NSW, 2052, Australia.
| | - Justin G Lees
- School of Medical Sciences, University of New South Wales, UNSW Medicine, Sydney, NSW, 2052, Australia.
| | - Cristina F Kim
- School of Medical Sciences, University of New South Wales, UNSW Medicine, Sydney, NSW, 2052, Australia.
| | - Barbara Cameron
- Centre for Infection and Inflammation Research, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Vasso Apostolopoulos
- College of Health and Biomedicine, Centre for Chronic Disease Prevention and Management, Victoria University, Melbourne, VIC, Australia.
| | - Gila Moalem-Taylor
- School of Medical Sciences, University of New South Wales, UNSW Medicine, Sydney, NSW, 2052, Australia.
| |
Collapse
|
89
|
The up-regulation of spinal Toll-like receptor 4 in rats with inflammatory pain induced by complete Freund's adjuvant. Brain Res Bull 2015; 111:97-103. [DOI: 10.1016/j.brainresbull.2015.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/22/2014] [Accepted: 01/05/2015] [Indexed: 11/23/2022]
|
90
|
Old EA, Clark AK, Malcangio M. The role of glia in the spinal cord in neuropathic and inflammatory pain. Handb Exp Pharmacol 2015; 227:145-170. [PMID: 25846618 DOI: 10.1007/978-3-662-46450-2_8] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Chronic pain, both inflammatory and neuropathic, is a debilitating condition in which the pain experience persists after the painful stimulus has resolved. The efficacy of current treatment strategies using opioids, NSAIDS and anticonvulsants is limited by the extensive side effects observed in patients, underlining the necessity for novel therapeutic targets. Preclinical models of chronic pain have recently provided evidence for a critical role played by glial cells in the mechanisms underlying the chronicity of pain, both at the site of damage in the periphery and in the dorsal horn of the spinal cord. Here microglia and astrocytes respond to the increased input from the periphery and change morphology, increase in number and release pro-nociceptive mediators such as ATP, cytokines and chemokines. These gliotransmitters can sensitise neurons by activation of their cognate receptors thereby contributing to central sensitization which is fundamental for the generation of allodynia, hyperalgesia and spontaneous pain.
Collapse
Affiliation(s)
- Elizabeth Amy Old
- Wolfson Centre for Age Related Diseases, King's College London, London, UK
| | | | | |
Collapse
|
91
|
Man'kovskaya YP, Maznychenko AV, Pil'kevych NO, Maisky VO, Vlasenko OV, Dovgan OV. [NADPH-DIAPHORASE REACTIVITY IN THE VENTRAL HORN OF THE FELINE SPINAL CORD DURING ACUTE MUSCLE INFLAMMATION]. ACTA ACUST UNITED AC 2015; 61:46-52. [PMID: 27025044 DOI: 10.15407/fz61.06.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of this research was to reveal the changes in the NADPH-d reactivity in the lumbal spinal cord (L6/L7) of cats with unilateral acute myositis of the mm. gastrocnemius-soleus after intramuscular injections of carrageenan. The effect of unilateral muscle inflammation was expressed in a significant increase in the number of NADPH-d-reactive neurons in ipsilateral and contralateral intermediate (lamina VII; 17.62 ± 2.7 and 20.67 ± 13.3) and medial (lamina VIII; 7.3 ± 1.9 and 6.0 ± 2.1 respectively) zones of the ventral horns. However, a clear decline of the reactive cells was recorded on the ipsilateral side within the area around the central canal (lamina X). An increase in the NADPH-d reactivity within the ventral horns on both sides on the spinal cord and the induction of such reactivity (contralaterally) in large multipolar neurons localized in the dorsal part of the intermediate zone were revealed in cats with unilateral acute muscle inflammation. It is hypothesized, that during acute myositis, plastic changes in different layers of the dorsal and ventral horns activate the processes of disinhibition due to an increase in the number of NOS-containing/NADPH-d-reactive neurons in the spinal gray matter.
Collapse
|
92
|
Involvement of pro- and antinociceptive factors in minocycline analgesia in rat neuropathic pain model. J Neuroimmunol 2014; 277:57-66. [PMID: 25304927 DOI: 10.1016/j.jneuroim.2014.09.020] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 01/08/2023]
Abstract
In neuropathic pain the repeated minocycline treatment inhibited the mRNA and protein expression of the microglial markers and metalloproteinase-9 (MMP-9). The minocycline diminished the pronociceptive (IL-6, IL-18), but not antinociceptive (IL-1alpha, IL-4, IL-10) cytokines at the spinal cord level. In vitro primary cell culture studies have shown that MMP-9, TIMP-1, IL-1beta, IL-1alpha, IL-6, IL-10, and IL-18 are of microglial origin. Minocycline reduces the production of pronociceptive factors, resulting in a more potent antinociceptive effect. This change in the ratio between pro- and antinociceptive factors, in favour of the latter may be the mechanism of minocycline analgesia in neuropathy.
Collapse
|
93
|
Xiang Y, Liu T, Yang H, Gao F, Xiang H, Manyande A, Tian Y, Tian X. NRG1-ErbB signalling promotes microglia activation contributing to incision-induced mechanical allodynia. Eur J Pain 2014; 19:686-94. [PMID: 25159022 DOI: 10.1002/ejp.590] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2014] [Indexed: 01/23/2023]
Affiliation(s)
- Y Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | | | | | | | | | |
Collapse
|
94
|
König C, Zharsky M, Möller C, Schaible HG, Ebersberger A. Involvement of peripheral and spinal tumor necrosis factor α in spinal cord hyperexcitability during knee joint inflammation in rats. Arthritis Rheumatol 2014; 66:599-609. [PMID: 24574220 DOI: 10.1002/art.38271] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 11/05/2013] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Tumor necrosis factor α (TNFα) is produced not only in peripheral tissues, but also in the spinal cord. The purpose of this study was to address the potential of peripheral and spinal TNFα to induce and maintain spinal hyperexcitability, which is a hallmark of pain states in the joints during rheumatoid arthritis and osteoarthritis. METHODS In vivo recordings of the responses of spinal cord neurons to nociceptive knee input under normal conditions and in the presence of experimental knee joint inflammation were obtained in anesthetized rats. TNFα, etanercept, or antibodies to TNF receptors were applied to either the knee joint or the spinal cord surface. RESULTS Injection of TNFα into the knee joint cavity increased the responses of spinal cord neurons to mechanical joint stimulation, and injection of etanercept into the knee joint reduced the inflammation-evoked spinal activity. These spinal effects closely mirrored the induction and reduction of peripheral sensitization. Responses to joint stimulation were also enhanced by spinal application of TNFα, and spinal application of either etanercept or anti-TNF receptor type I significantly attenuated the generation of inflammation-evoked spinal hyperexcitability, which is characterized by widespread pain sensitization beyond the inflamed joint. Spinally applied etanercept did not reduce established hyperexcitability in the acute kaolin/carrageenan model. In antigen-induced arthritis, etanercept decreased spinal responses on day 1, but not on day 3. CONCLUSION While peripheral TNFα increases spinal responses to joint stimulation, spinal TNFα supports the generation of the full pattern of spinal hyperexcitability. However, established spinal hyperexcitability may be maintained by downstream mechanisms that are independent of spinal TNFα.
Collapse
Affiliation(s)
- Christian König
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | | | | | | | | |
Collapse
|
95
|
The pivotal role played by lipocalin-2 in chronic inflammatory pain. Exp Neurol 2014; 254:41-53. [PMID: 24440229 DOI: 10.1016/j.expneurol.2014.01.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 12/13/2013] [Accepted: 01/07/2014] [Indexed: 12/30/2022]
|
96
|
Chen W, Walwyn W, Ennes HS, Kim H, McRoberts JA, Marvizón JCG. BDNF released during neuropathic pain potentiates NMDA receptors in primary afferent terminals. Eur J Neurosci 2014; 39:1439-54. [PMID: 24611998 DOI: 10.1111/ejn.12516] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/13/2014] [Accepted: 01/16/2014] [Indexed: 11/28/2022]
Abstract
NMDA receptors in primary afferent terminals can contribute to hyperalgesia by increasing neurotransmitter release. In rats and mice, we found that the ability of intrathecal NMDA to induce neurokinin 1 receptor (NK1R) internalization (a measure of substance P release) required a previous injection of BDNF. Selective knock-down of NMDA receptors in primary afferents decreased NMDA-induced NK1R internalization, confirming the presynaptic location of these receptors. The effect of BDNF was mediated by tropomyosin-related kinase B (trkB) receptors and not p75 neurotrophin receptors (p75(NTR) ), because it was not produced by proBDNF and was inhibited by the trkB antagonist ANA-12 but not by the p75(NTR) inhibitor TAT-Pep5. These effects are probably mediated through the truncated form of the trkB receptor as there is little expression of full-length trkB in dorsal root ganglion (DRG) neurons. Src family kinase inhibitors blocked the effect of BDNF, suggesting that trkB receptors promote the activation of these NMDA receptors by Src family kinase phosphorylation. Western blots of cultured DRG neurons revealed that BDNF increased Tyr(1472) phosphorylation of the NR2B subunit of the NMDA receptor, known to have a potentiating effect. Patch-clamp recordings showed that BDNF, but not proBDNF, increased NMDA receptor currents in cultured DRG neurons. NMDA-induced NK1R internalization was also enabled in a neuropathic pain model or by activating dorsal horn microglia with lipopolysaccharide. These effects were decreased by a BDNF scavenger, a trkB receptor antagonist and a Src family kinase inhibitor, indicating that BDNF released by microglia potentiates NMDA receptors in primary afferents during neuropathic pain.
Collapse
Affiliation(s)
- Wenling Chen
- Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA; Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
97
|
Le Coz GM, Fiatte C, Anton F, Hanesch U. Differential neuropathic pain sensitivity and expression of spinal mediators in Lewis and Fischer 344 rats. BMC Neurosci 2014; 15:35. [PMID: 24575861 PMCID: PMC3975939 DOI: 10.1186/1471-2202-15-35] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/25/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Altered hypothalamo-pituitary-adrenal (HPA) axis activity may be accompanied by a modulation of pain sensitivity. In a model of neuropathic pain (chronic constriction injury, CCI) we investigated the onset and maintenance of mechanical allodynia/hyperalgesia and the expression of biochemical mediators potentially involved in spinal cell modulation in two rat strains displaying either hypo- (Lewis-LEW) or hyper- (Fischer 344-FIS) reactivity of the HPA axis. RESULTS Mechanical pain thresholds and plasmatic corticosterone levels were assessed before and during periods of 4 or 21 days following CCI surgery. At the end of the respective protocols, the mRNA expression of glial cell markers (GFAP and Iba1) and glutamate transporters (EAAT3 and EAAT2) were examined. We observed a correlation between the HPA axis reactivity and the pain behavior but not as commonly described in the literature; LEW rats seemed to be less sensitive than FIS from 4 to 14 days after the CCI surgery when looking at the mechanical allodynia/hyperalgesia. However, the biochemical spinal markers expression we observed is conflicting. CONCLUSION We did not find a specific causal relation between the pain behavior and the glial cell activation or the expression of the glutamate transporters, suggesting that the interaction between the HPA axis and the spinal activation pattern is more complex in a context of neuropathic pain.
Collapse
Affiliation(s)
| | | | | | - Ulrike Hanesch
- Laboratory of Neurophysiology & Psychobiology, University of Luxembourg, 162a, avenue de la Faïencerie, Luxembourg, L-1511, Luxembourg.
| |
Collapse
|
98
|
Li KW, Yu YP, Zhou C, Kim DS, Lin B, Sharp K, Steward O, Luo ZD. Calcium channel α2δ1 proteins mediate trigeminal neuropathic pain states associated with aberrant excitatory synaptogenesis. J Biol Chem 2014; 289:7025-7037. [PMID: 24459143 DOI: 10.1074/jbc.m114.548990] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To investigate a potential mechanism underlying trigeminal nerve injury-induced orofacial hypersensitivity, we used a rat model of chronic constriction injury to the infraorbital nerve (CCI-ION) to study whether CCI-ION caused calcium channel α2δ1 (Cavα2δ1) protein dysregulation in trigeminal ganglia and associated spinal subnucleus caudalis and C1/C2 cervical dorsal spinal cord (Vc/C2). Furthermore, we studied whether this neuroplasticity contributed to spinal neuron sensitization and neuropathic pain states. CCI-ION caused orofacial hypersensitivity that correlated with Cavα2δ1 up-regulation in trigeminal ganglion neurons and Vc/C2. Blocking Cavα2δ1 with gabapentin, a ligand for the Cavα2δ1 proteins, or Cavα2δ1 antisense oligodeoxynucleotides led to a reversal of orofacial hypersensitivity, supporting an important role of Cavα2δ1 in orofacial pain processing. Importantly, increased Cavα2δ1 in Vc/C2 superficial dorsal horn was associated with increased excitatory synaptogenesis and increased frequency, but not the amplitude, of miniature excitatory postsynaptic currents in dorsal horn neurons that could be blocked by gabapentin. Thus, CCI-ION-induced Cavα2δ1 up-regulation may contribute to orofacial neuropathic pain states through abnormal excitatory synapse formation and enhanced presynaptic excitatory neurotransmitter release in Vc/C2.
Collapse
Affiliation(s)
- Kang-Wu Li
- Department of Anesthesiology and Perioperative Care, University of California School of Medicine, Irvine, California 92697
| | - Yanhui Peter Yu
- Department of Pharmacology, University of California School of Medicine, Irvine, California 92697
| | - Chunyi Zhou
- Department of Pharmacology, University of California School of Medicine, Irvine, California 92697
| | - Doo-Sik Kim
- Department of Anesthesiology and Perioperative Care, University of California School of Medicine, Irvine, California 92697
| | - Bin Lin
- Department of Anesthesiology and Perioperative Care, University of California School of Medicine, Irvine, California 92697
| | - Kelli Sharp
- Reeve-Irvine Research Center, University of California School of Medicine, Irvine, California 92697
| | - Oswald Steward
- Reeve-Irvine Research Center, University of California School of Medicine, Irvine, California 92697
| | - Z David Luo
- Department of Anesthesiology and Perioperative Care, University of California School of Medicine, Irvine, California 92697; Department of Pharmacology, University of California School of Medicine, Irvine, California 92697; Reeve-Irvine Research Center, University of California School of Medicine, Irvine, California 92697.
| |
Collapse
|
99
|
Jeon S, Jha MK, Ock J, Seo J, Jin M, Cho H, Lee WH, Suk K. Role of lipocalin-2-chemokine axis in the development of neuropathic pain following peripheral nerve injury. J Biol Chem 2013; 288:24116-27. [PMID: 23836894 DOI: 10.1074/jbc.m113.454140] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Lipocalin 2 (LCN2), which is also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), binds small, hydrophobic ligands and interacts with cell surface receptor 24p3R to regulate diverse cellular processes. In the present study, we examined the role of LCN2 in the pathogenesis of neuropathic pain using a mouse model of spared nerve injury (SNI). Lcn2 mRNA levels were significantly increased in the dorsal horn of the spinal cord after SNI, and LCN2 protein was mainly localized in neurons of the dorsal and ventral horns. LCN2 receptor 24p3R was expressed in spinal neurons and microglia after SNI. Lcn2-deficient mice exhibited significantly less mechanical pain hypersensitivity during the early phase after SNI, and an intrathecal injection of recombinant LCN2 protein elicited mechanical pain hypersensitivity in naive animals. Lcn2 deficiency, however, did not affect acute nociceptive pain. Lcn2-deficient mice showed significantly less microglial activation and proalgesic chemokine (CCL2 and CXCL1) production in the spinal cord after SNI than wild-type mice, and recombinant LCN2 protein induced the expression of these chemokines in cultured neurons. Furthermore, the expression of LCN2 and its receptor was detected in neutrophils and macrophages in the sciatic nerve following SNI, suggesting the potential role of peripheral LCN2 in neuropathic pain. Taken together, our results indicate that LCN2 plays a critical role in the development of pain hypersensitivity following peripheral nerve injury and suggest that LCN2 mediates neuropathic pain by inducing chemokine expression and subsequent microglial activation.
Collapse
Affiliation(s)
- Sangmin Jeon
- Department of Pharmacology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Different peripheral tissue injury induces differential phenotypic changes of spinal activated microglia. Clin Dev Immunol 2013; 2013:901420. [PMID: 23818916 PMCID: PMC3681311 DOI: 10.1155/2013/901420] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 05/04/2013] [Accepted: 05/16/2013] [Indexed: 01/23/2023]
Abstract
The purpose of this study is to investigate the possible different cellular marker expression associated with spinal cord microglial activation in different pain models. Immunohistochemistry and western blotting analysis of CD45, CD68, and MHC class I antigen as well as CD11b and Iba-1 in the spinal cord were quantitatively compared among widely used three pain animal models, complete Freund's adjuvant (CFA) injection, formalin injection, and chronic constriction injury (CCI) models. The results showed that significant upregulated expressions of CD45 and MHC class I antigen in spinal microglia as well as morphological changes with increased staining with CD11b and Iba-1 were seen in CCI and formalin models and not found in CFA-induced inflammatory pain model. CD68 expression was only detected in CCI model. Our findings suggested that different peripheral tissue injuries produced differential phenotypic changes associated with spinal microglial activation; peripheral nerve injury might induce spinal microglia to acquire these immunomolecular phenotypic changes.
Collapse
|