301
|
Vicario N, Parenti R, Arico' G, Turnaturi R, Scoto GM, Chiechio S, Parenti C. Repeated activation of delta opiod receptors counteracts nerve injury-induced TNF-α up-regulation in the sciatic nerve of rats with neuropathic pain: A possible correlation with delta opiod receptors-mediated antiallodinic effect. Mol Pain 2016; 12:12/0/1744806916667949. [PMID: 27590071 PMCID: PMC5024981 DOI: 10.1177/1744806916667949] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Despite mu opioid receptor agonists are the cornerstones of moderate-to-severe acute pain treatment, their effectiveness in chronic pain conditions is controversial. In contrast to mu opioid receptor agonists, a number of studies have reported the effectiveness of delta opioid receptor agonists on neuropathic pain strengthening the idea that delta opioid receptors gain importance when chronic pain develops. Among other effects, it has been shown that delta opioid receptor activation in optic nerve astrocytes inhibits tumor necrosis factor-α-mediated inflammation in response to severe hypoxia. Considering the involvement of tumor necrosis factor-α in the development and maintenance of neuropathic pain, with this study we sought to correlate the effect of delta opioid receptor agonist on the development of mechanical allodynia to tumor necrosis factor-α expression at the site of nerve injury in rats subjected to chronic constriction injury of the sciatic nerve. To this aim, we measured the levels of tumor necrosis factor-α in the sciatic nerve of rats with neuropathic pain after repeated injections with a delta opioid receptor agonist. Results obtained demonstrated that repeated administrations of the delta opioid receptor agonist SNC80 (10 mg/kg, i.p. for seven consecutive days) significantly inhibited the development of mechanical allodynia in rats with neuropathic pain and that the improvement of neuropathic symptom was timely related to the reduced expression of tumor necrosis factor-α in the rat sciatic nerve. We demonstrated also that when treatment with the delta opioid receptor agonist was suspended both allodynia and tumor necrosis factor-α up-regulation in the sciatic nerve of rats with neuropathic pain were restored. These results show that persistent delta opioid receptor activation significantly attenuates neuropathic pain and negatively regulates sciatic nerve tumor necrosis factor-α expression in chronic constriction injury rats.
Collapse
Affiliation(s)
| | | | | | | | | | - Santina Chiechio
- University of CataniaUniversity of CataniaUniversity of CataniaUniversity of CataniaUniversity of Catania
| | - Carmela Parenti
- University of CataniaUniversity of CataniaUniversity of CataniaUniversity of CataniaUniversity of Catania
| |
Collapse
|
302
|
Cooper MA, Kluding PM, Wright DE. Emerging Relationships between Exercise, Sensory Nerves, and Neuropathic Pain. Front Neurosci 2016; 10:372. [PMID: 27601974 PMCID: PMC4993768 DOI: 10.3389/fnins.2016.00372] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/02/2016] [Indexed: 01/09/2023] Open
Abstract
The utilization of physical activity as a therapeutic tool is rapidly growing in the medical community and the role exercise may offer in the alleviation of painful disease states is an emerging research area. The development of neuropathic pain is a complex mechanism, which clinicians and researchers are continually working to better understand. The limited therapies available for alleviation of these pain states are still focused on pain abatement and as opposed to treating underlying mechanisms. The continued research into exercise and pain may address these underlying mechanisms, but the mechanisms which exercise acts through are still poorly understood. The objective of this review is to provide an overview of how the peripheral nervous system responds to exercise, the relationship of inflammation and exercise, and experimental and clinical use of exercise to treat pain. Although pain is associated with many conditions, this review highlights pain associated with diabetes as well as experimental studies on nerve damages-associated pain. Because of the global effects of exercise across multiple organ systems, exercise intervention can address multiple problems across the entire nervous system through a single intervention. This is a double-edged sword however, as the global interactions of exercise also require in depth investigations to include and identify the many changes that can occur after physical activity. A continued investment into research is necessary to advance the adoption of physical activity as a beneficial remedy for neuropathic pain. The following highlights our current understanding of how exercise alters pain, the varied pain models used to explore exercise intervention, and the molecular pathways leading to the physiological and pathological changes following exercise intervention.
Collapse
Affiliation(s)
- Michael A Cooper
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City, KS, USA
| | - Patricia M Kluding
- Department of Physical Therapy and Rehabilitation Science, University of Kansas Medical Center Kansas City, KS, USA
| | - Douglas E Wright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center Kansas City, KS, USA
| |
Collapse
|
303
|
Chen NF, Chen WF, Sung CS, Lu CH, Chen CL, Hung HC, Feng CW, Chen CH, Tsui KH, Kuo HM, Wang HMD, Wen ZH, Huang SY. Contributions of p38 and ERK to the antinociceptive effects of TGF-β1 in chronic constriction injury-induced neuropathic rats. J Headache Pain 2016; 17:72. [PMID: 27541934 PMCID: PMC4991976 DOI: 10.1186/s10194-016-0665-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022] Open
Abstract
Background Transforming growth factor-βs (TGF-βs) are a group of multifunctional proteins that have neuroprotective roles in various experimental models. We previously reported that intrathecal (i.t.) injections of TGF-β1 significantly inhibit neuropathy-induced thermal hyperalgesia, spinal microglia and astrocyte activation, as well as upregulation of tumor necrosis factor-α. However, additional cellular mechanisms for the antinociceptive effects of TGF-β1, such as the mitogen-activated protein kinase (MAPK) pathway, have not been elucidated. During persistent pain, activation of MAPKs, especially p38 and extracellular signal-regulated kinase (ERK), have crucial roles in the induction and maintenance of pain hypersensitivity, via both nontranscriptional and transcriptional regulation. In the present study, we used a chronic constriction injury (CCI) rat model to explore the role of spinal p38 and ERK in the analgesic effects of TGF-β1. Methods We investigated the cellular mechanisms of the antinociceptive effects of i.t. injections of TGF-β1 in CCI induced neuropathic rats by spinal immunohistofluorescence analyses. Results The results demonstrated that the antinociceptive effects of TGF-β1 (5 ng) were maintained at greater than 50 % of the maximum possible effect in rats with CCI for at least 6 h after a single i.t. administration. Thus, we further examined these alterations in spinal p38 and ERK from 0.5 to 6 h after i.t. administration of TGF-β1. TGF-β1 significantly attenuated CCI-induced upregulation of phosphorylated p38 (phospho-p38) and phosphorylated ERK (phospho-ERK) expression in the dorsal horn of the lumbar spinal cord. Double immunofluorescence staining illustrated that upregulation of spinal phospho-p38 was localized to neurons, activated microglial cells, and activated astrocytes in rats with CCI. Additionally, increased phospho-ERK occurred in activated microglial cells and activated astrocytes. Furthermore, i.t. administration of TGF-β1 markedly inhibited phospho-p38 upregulation in neurons, microglial cells, and astrocytes. However, i.t. injection of TGF-β1 also reduced phospho-ERK upregulation in microglial cells and astrocytes. Conclusions The present results demonstrate that suppressing p38 and ERK activity affects TGF-β1-induced analgesia during neuropathy.
Collapse
Affiliation(s)
- Nan-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan.,Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan.,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan.,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, 11217, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Ching-Hsiang Lu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan
| | - Chun-Lin Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan
| | - Han-Chun Hung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 80424, Taiwan
| | - Chien-Wei Feng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 80424, Taiwan
| | - Chun-Hong Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 80424, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan.,Department of Obstetrics and Gynecology and Institute of Clinical Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.,Department of Pharmacy and Graduate Institute of Pharmaceutical Technology, Tajen University, Pingtung, 90741, Taiwan
| | - Hsiao-Mei Kuo
- Center for Neuroscience, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 40227, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.,College of Oceanology and Food Scienece, Quanzhou Normal University, Quanzhou, 362000, China
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 80424, Taiwan. .,Marine Biomedical Laboratory and Center for Translational Biopharmaceuticals, Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.
| | - Shi-Ying Huang
- Center for Neuroscience, National Sun Yat-sen University, #70 Lien-Hai Rd, Kaohsiung, 80424, Taiwan. .,College of Oceanology and Food Scienece, Quanzhou Normal University, Quanzhou, 362000, China.
| |
Collapse
|
304
|
Targeting the innate repair receptor to treat neuropathy. Pain Rep 2016; 1:e566. [PMID: 29392190 PMCID: PMC5741312 DOI: 10.1097/pr9.0000000000000566] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/13/2016] [Accepted: 06/13/2016] [Indexed: 11/26/2022] Open
Abstract
The innate repair receptor (IRR) is a heteromer of the erythropoietin receptor and the β-common (CD131) receptor, which simultaneously activates anti-inflammatory and tissue repair pathways. Experimental data suggest that after peripheral nerve injury, the IRR is upregulated in the spinal cord and modulates the neurogenic inflammatory response. The recently introduced selective IRR agonist ARA290 is an 11-amino acid peptide initially tested in animal models of neuropathy. After sciatic nerve injury, ARA290 produced a rapid and long-term relief of mechanical and cold allodynia in normal mice, but not in animals with a β-common receptor knockout phenotype. In humans, ARA290 has been evaluated in patients with small fiber neuropathy associated with sarcoidosis or type 2 diabetes (T2D) mellitus. In patients with sarcoidosis, ARA290 significantly improved neuropathic and autonomic symptoms, as well as quality of life as assessed by the small fiber neuropathy screening list questionnaire. In addition, ARA290 treatment for 28 days initiated a regrowth of small nerve fibers in the cornea, but not in the epidermis. In patients with T2D, the results were similar to those observed in patients with sarcoidosis along with an improved metabolic profile. In both populations, ARA290 lacked significant adverse effects. These experimental and clinical studies show that ARA290 effectively reprograms a proinflammatory, tissue-damaging milieu into one of healing and tissue repair. Further clinical trials with long-term treatment and follow-up are needed to assess the full potential of IRR activation by ARA290 as a disease-modifying therapy in neuropathy of various etiologies.
Collapse
|
305
|
Yang F, Peng L, Luo J, Yi H, Hu X. Intra-amygdala microinfusion of neuropeptide S attenuates neuropathic pain and suppresses the response of spinal microglia and astrocytes after spinal nerve ligation in rats. Peptides 2016; 82:26-34. [PMID: 27224019 DOI: 10.1016/j.peptides.2016.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 05/13/2016] [Accepted: 05/15/2016] [Indexed: 01/04/2023]
Abstract
The amygdala circuitry and neuropeptide S (NPS) have been shown to play an important role in the pain modulation. However, the alleviative effect of NPS in amygdala on neuropathic pain (NP) is not fully understood. Here, we demonstrate a possibility that the intra-amygdala microinfusion of NPS attenuates NP symptoms and suppresses the response of spinal microglia and astrocytes after spinal nerve injury. Spinal nerve ligation (SNL) in rats resulted in a striking decline in level of NPS and density of NPS-immunopositive cells in amygdala. SNL rats randomly received chronic bilateral microinjections of NPS (1, 10 and 100pmol/side) or saline into the amygdala via cannulas on days 3, 6, 9, 12, 15 and 18 post-surgery. Chronic treatment with NPS increased thermal withdrawal latency (TWL) and mechanical withdrawal threshold (MWT) on day 11-21 post-SNL. The simultaneous treatment with SHA68 as non-peptide NPS receptor antagonist decreased the TWL and MWT, and reversed the inhibitory effects of NPS in SNL rats. NPS also significantly attenuated immunoreactivities of ionized calcium-binding adapter molecule 1 and glial fibrillary acidic protein for microglia and astrocytes. Furthermore, the elevated levels of inflammatory mediators and expressions of nuclear factor κB p65 and CX3C chemokine receptor 1 due to SNL were significantly attenuated by NPS in amygdala. These effects of NPS were also counteracted by SHA 68. SHA 68 per se deteriorated the symptom of NP and the response of spinal microglia and astrocytes in SNL rats. Our study identified a protective role for NPS in amygdala against the development of NP, possibly attributing to its anti-inflammatory activity and inhibition of spinal microglia and astrocytes.
Collapse
Affiliation(s)
- Fengrui Yang
- Department of Anesthesiology, the First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Liangyu Peng
- Department of Anesthesiology, the First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jingjing Luo
- Department of Anesthesiology, the First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Han Yi
- Department of Anesthesiology, the First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Xiaoling Hu
- Department of Anesthesiology, the First Affiliated Hospital of University of South China, Hengyang 421001, China.
| |
Collapse
|
306
|
Kremer M, Salvat E, Muller A, Yalcin I, Barrot M. Antidepressants and gabapentinoids in neuropathic pain: Mechanistic insights. Neuroscience 2016; 338:183-206. [PMID: 27401055 DOI: 10.1016/j.neuroscience.2016.06.057] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/21/2016] [Accepted: 06/30/2016] [Indexed: 01/20/2023]
Abstract
Neuropathic pain arises as a consequence of a lesion or disease affecting the somatosensory system. It is generally chronic and challenging to treat. The recommended pharmacotherapy for neuropathic pain includes the use of some antidepressants, such as tricyclic antidepressants (TCAs) (amitriptyline…) or serotonin and noradrenaline re-uptake inhibitors (duloxetine…), and/or anticonvulsants such as the gabapentinoids gabapentin or pregabalin. Antidepressant drugs are not acute analgesics but require a chronic treatment to relieve neuropathic pain, which suggests the recruitment of secondary downstream mechanisms as well as long-term molecular and neuronal plasticity. Noradrenaline is a major actor for the action of antidepressant drugs in a neuropathic pain context. Mechanistic hypotheses have implied the recruitment of noradrenergic descending pathways as well as the peripheral recruitment of noradrenaline from sympathetic fibers sprouting into dorsal root ganglia; and importance of both α2 and β2 adrenoceptors have been reported. These monoamine re-uptake inhibitors may also indirectly act as anti-proinflammatory cytokine drugs; and their therapeutic action requires the opioid system, particularly the mu (MOP) and/or delta (DOP) opioid receptors. Gabapentinoids, which target the voltage-dependent calcium channels α2δ-1 subunit, inhibit calcium currents, thus decreasing the excitatory transmitter release and spinal sensitization. Gabapentinoids also activate the descending noradrenergic pain inhibitory system coupled to spinal α2 adrenoceptors. Gabapentinoid treatment may also indirectly impact on neuroimmune actors, like proinflammatory cytokines. These drugs are effective against neuropathic pain both with acute administration at high dose and with repeated administration. This review focuses on mechanistic knowledge concerning chronic antidepressant treatment and gabapentinoid treatment in a neuropathic pain context.
Collapse
Affiliation(s)
- Mélanie Kremer
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | - Eric Salvat
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Centre d'Etude et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - André Muller
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Centre d'Etude et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Michel Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France.
| |
Collapse
|
307
|
Kalliomäki ML, Sandblom G, Hallberg M, Grönbladh A, Gunnarsson U, Gordh T, Ginya H, Nyberg F. Genetic susceptibility to postherniotomy pain. The influence of polymorphisms in the Mu opioid receptor, TNF-α, GRIK3, GCH1, BDNF and CACNA2D2 genes. Scand J Pain 2016; 12:1-6. [PMID: 28850479 DOI: 10.1016/j.sjpain.2015.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/14/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS Despite improvements in surgical technique, 5%-8% of patients undergoing herniorrhaphy still suffer from clinically relevant persistent postherniotomy pain. This is a problem at both individual and society levels. The aim of this study was to determine whether or not a single nucleotide polymorphism in a specific gene contributes to the development of persistent pain after surgery. METHODS One hundred individuals with persistent postherniotomy pain, along with 100 without pain matched for age, gender and type of surgery were identified in a previous cohort study on patients operated for groin hernia. All patients underwent a thorough sensory examination and blood samples were collected. DNA was extracted and analysed for single nucleotide polymorphism in the Mu opioid receptor, TNF-α, GRIK3, GCH1, BDNF and CACNA2D2 genes. RESULTS Patients with neuropathic pain were found to have a homozygous single nucleotide polymorph in the TNF-α gene significantly more often than pain-free patients (P=0.036, one-tailed test). CONCLUSIONS SNP in the TNF-α gene has a significant impact on the risk for developing PPSP. IMPLICATIONS The result suggests the involvement of genetic variance in the development of pain and this requires further investigation.
Collapse
Affiliation(s)
- Maija-Liisa Kalliomäki
- Uppsala University, Department for Surgical Sciences, Uppsala, Sweden; Department of Anaesthesia, Tampere University Hospital, Finland.
| | | | - Mathias Hallberg
- Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, Uppsala, Sweden
| | - Alfhild Grönbladh
- Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, Uppsala, Sweden
| | - Ulf Gunnarsson
- Department of Surgical and Perioperative Sciences, Umeå University, Sweden
| | - Torsten Gordh
- Uppsala University, Department for Surgical Sciences, Uppsala, Sweden; Pain Centre, Uppsala University Hospital, Uppsala, Sweden
| | - Harumi Ginya
- Division of IVD System Development, Precision System Science Co., Ltd., Chiba, Japan
| | - Fred Nyberg
- Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, Uppsala, Sweden
| |
Collapse
|
308
|
Nakanishi M, Nakae A, Kishida Y, Baba K, Sakashita N, Shibata M, Yoshikawa H, Hagihara K. Go-sha-jinki-Gan (GJG) ameliorates allodynia in chronic constriction injury-model mice via suppression of TNF-α expression in the spinal cord. Mol Pain 2016; 12:12/0/1744806916656382. [PMID: 27296622 PMCID: PMC4956397 DOI: 10.1177/1744806916656382] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background Alternative medicine is noted for its clinical effect and minimal invasiveness in the treatment of neuropathic pain. Go-sha-jinki-Gan, a traditional Japanese herbal medicine, has been used for meralgia and numbness in elderly patients. However, the exact mechanism of GJG is unclear. This study aimed to investigate the molecular mechanism of the analgesic effect of GJG in a chronic constriction injury model. Results GJG significantly reduced allodynia and hyperalgesia from the early phase (von Frey test, p < 0.0001; cold-plate test, p < 0.0001; hot-plate test p = 0.011; two-way repeated measures ANOVA). Immunohistochemistry and Western blot analysis revealed that GJG decreased the expression of Iba1 and tumor necrosis factor-α in the spinal cord. Double staining immunohistochemistry showed that most of the tumor necrosis factor-α was co-expressed in Iba1-positive cells at day 3 post-operation. GJG decreased the phosphorylation of p38 in the ipsilateral dorsal horn. Moreover, intrathecal injection of tumor necrosis factor-α opposed the anti-allodynic effect of GJG in the cold-plate test. Conclusions Our data suggest that GJG ameliorates allodynia in chronic constriction injury model mice via suppression of tumor necrosis factor-α expression derived from activated microglia. GJG is a promising drug for the treatment of neuropathic pain induced by neuro-inflammation.
Collapse
Affiliation(s)
| | - Aya Nakae
- Osaka University Graduate School of medicineOsaka University Graduate School of medicineOsaka University Graduate School of medicineOsaka University Graduate School of medicine Osaka University Graduate School of Medicine Osaka University Graduate School of Medicine
| | | | | | | | | | | | - Keisuke Hagihara
- Osaka University Graduate School of medicineOsaka University Graduate School of medicineOsaka University Graduate School of medicineOsaka University Graduate School of medicine Osaka University Graduate School of Medicine Osaka University Graduate School of Medicine
| |
Collapse
|
309
|
Al Salihi MO, Kobayashi M, Tamari K, Miyamura T, Takeuchi K. Tumor necrosis factor-α antagonist suppresses local inflammatory reaction and facilitates olfactory nerve recovery following injury. Auris Nasus Larynx 2016; 44:70-78. [PMID: 27297522 DOI: 10.1016/j.anl.2016.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 04/18/2016] [Accepted: 05/13/2016] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Olfactory dysfunction is a common finding in head trauma due to injury to the olfactory nerve. We previously reported that anti-inflammatory treatment with steroids improves recovery outcome in olfactory nerve injury models. Clinically, however, steroid administration is not recommended in the acute phase of head injury cases because of concerns regarding its side effects. Tumor necrosis factor (TNF-α) is known to play a key role in inflammatory response to injury. The present study examines if the inhibition of TNF-α can facilitate functional recovery in the olfactory system following injury. MATERIALS AND METHODS Olfactory nerve transection (NTx) was performed in olfactory marker protein (OMP-tau-lacZ) mice to establish injury models. We measured TNF-α gene expression in the olfactory bulb using semi-quantitative and real time polymerase chain reaction (PCR) assays and found that they increase within hours after NTx injury. A TNF-α antagonist (etanercept) was intraperitoneally injected immediately after the NTx and histological assessment of recovery within the olfactory bulb was performed at 5-70 days. X-gal staining labeled OMP in the degenerating and regenerating olfactory nerve fibers, and immunohistochemical staining detected the presence of reactive astrocytes and macrophages/microglia. RESULTS Etanercept-injected mice showed significantly smaller areas of injury-associated tissue, fewer astrocytes and macrophages/microglia, and an increase in regenerating nerve fibers. Olfactory function assessments using both an olfactory avoidance behavioral test and evoked potential recordings showed improved functional recovery in etanercept-injected animals. CONCLUSION These findings suggest that inhibition of TNF-α could provide a new therapeutic strategy for the treatment of olfactory dysfunction following head injuries.
Collapse
Affiliation(s)
- Mohammed Omar Al Salihi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Masayoshi Kobayashi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan.
| | - Kengo Tamari
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Tomotaka Miyamura
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| |
Collapse
|
310
|
Karras S, Rapti E, Matsoukas S, Kotsa K. Vitamin D in Fibromyalgia: A Causative or Confounding Biological Interplay? Nutrients 2016; 8:nu8060343. [PMID: 27271665 PMCID: PMC4924184 DOI: 10.3390/nu8060343] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/18/2016] [Accepted: 05/26/2016] [Indexed: 01/13/2023] Open
Abstract
Fibromyalgia (FM) is a chronic syndrome with an increasing prevalence, characterized by widespread musculoskeletal pain in combination with a variety of cognitive symptoms and fatigue. A plethora of scientific evidence that has accumulated during the last decades, resulted in a significant improvement of the understanding of the pathophysiology of the disease. However, current therapeutic approaches in patients with FM remains a multidimensional approach including patient education, behavioral therapy, exercise, pain management, and relief of chronic symptoms, rather than the use drug therapies, based on the mechanisms of disease development. Vitamin D, a fat-soluble vitamin derived mainly from skin synthesis through ultraviolet radiation, has been recognized to manifest a plethora of extraskeletal actions, apart from its fundamental role in skeletal and calcium homeostasis, including modulation of cell growth, neuromuscular actions, and potential anti-inflammatory properties. Recent findings indicate that hypovitaminosis D to be highly prevalent in patients with FM. Supplementation studies are limited so far, indicating potential beneficial effects on pain and severity of the disease, however specific recommendations are lacking. This review aims to summarize and critically appraise data regarding the pathophysiological interplay between vitamin D and FM, available results from observational and supplementation studies so far, with a clinical discourse on current knowledge gaps and future research agenda.
Collapse
Affiliation(s)
- Spyridon Karras
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, AHEPA Hospital, Thessaloniki 54636, Greece.
| | - Eleni Rapti
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, AHEPA Hospital, Thessaloniki 54636, Greece.
| | - Stauros Matsoukas
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, AHEPA Hospital, Thessaloniki 54636, Greece.
| | - Kalliopi Kotsa
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, AHEPA Hospital, Thessaloniki 54636, Greece.
| |
Collapse
|
311
|
Rozas P, Lazcano P, Piña R, Cho A, Terse A, Pertusa M, Madrid R, Gonzalez-Billault C, Kulkarni AB, Utreras E. Targeted overexpression of tumor necrosis factor-α increases cyclin-dependent kinase 5 activity and TRPV1-dependent Ca2+ influx in trigeminal neurons. Pain 2016; 157:1346-1362. [PMID: 26894912 PMCID: PMC4868804 DOI: 10.1097/j.pain.0000000000000527] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We reported earlier that TNF-α, a proinflammatory cytokine implicated in many inflammatory disorders causing orofacial pain, increases the activity of Cdk5, a key kinase involved in brain development and function and recently found to be involved in pain signaling. To investigate a potential mechanism underlying inflammatory pain in trigeminal ganglia (TGs), we engineered a transgenic mouse model (TNF) that can conditionally overexpresses TNF-α upon genomic recombination by Cre recombinase. TNF mice were bred with Nav1.8-Cre mouse line that expresses the Cre recombinase in sensory neurons to obtain TNF-α:Nav1.8-Cre (TNF-α cTg) mice. Although TNF-α cTg mice appeared normal without any gross phenotype, they displayed a significant increase in TNF-α levels after activation of NFκB signaling in the TG. IL-6 and MCP-1 levels were also increased along with intense immunostaining for Iba1 and GFAP in TG, indicating the presence of infiltrating macrophages and the activation of satellite glial cells. TNF-α cTg mice displayed increased trigeminal Cdk5 activity, and this increase was associated with elevated levels of phospho-T407-TRPV1 and capsaicin-evocated Ca influx in cultured trigeminal neurons. Remarkably, this effect was prevented by roscovitine, an inhibitor of Cdk5, which suggests that TNF-α overexpression induced sensitization of the TRPV1 channel. Furthermore, TNF-α cTg mice displayed more aversive behavior to noxious thermal stimulation (45°C) of the face in an operant pain assessment device as compared with control mice. In summary, TNF-α overexpression in the sensory neurons of TNF-α cTg mice results in inflammatory sensitization and increased Cdk5 activity; therefore, this mouse model would be valuable for investigating the mechanism of TNF-α involved in orofacial pain.
Collapse
Affiliation(s)
- Pablo Rozas
- Laboratory of Molecular and Cellular Mechanisms of Pain, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Pablo Lazcano
- Laboratory of Molecular and Cellular Mechanisms of Pain, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Ricardo Piña
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Andrew Cho
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Anita Terse
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Maria Pertusa
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Rodolfo Madrid
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cellular and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Ashok B. Kulkarni
- Functional Genomics Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Elias Utreras
- Laboratory of Molecular and Cellular Mechanisms of Pain, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| |
Collapse
|
312
|
Chen Y, Zhang Y, Huo Y, Wang D, Hong Y. Adrenomedullin mediates tumor necrosis factor-α-induced responses in dorsal root ganglia in rats. Brain Res 2016; 1644:183-91. [PMID: 27184601 DOI: 10.1016/j.brainres.2016.05.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/30/2016] [Accepted: 05/11/2016] [Indexed: 01/08/2023]
Abstract
Adrenomedullin (AM), a member of the calcitonin gene-related peptide (CGRP) family, has been demonstrated to be a pain peptide. This study investigated the possible involvement of AM in tumor necrosis factor-alpha (TNF-α)-induced responses contributing to neuronal plasticity in the dorsal root ganglia (DRG). Exposure of the DRG explant cultures to TNF-α (5nM) for 48h upregulated the expression of AM mRNA. The treatment with TNF-α also increased the level of CGRP, CCL-2 and MMP-9 mRNA in the cultured DRG. This increase was attenuated by the co-treatment with the selective AM receptor antagonist AM22-52 (2μM). The blockade of AM receptors inhibited TNF-α-induced increase of the glial fibrillary acidic protein (GFAP), interleukin-1β (IL-1β), phosphorylated cAMP response element binding protein (pCREB) and nuclear factor kappa B (pNF-κB) proteins. On the other hand, the treatment with the AM receptor agonist AM1-50 (10nM) for 96h induced an increase in the level of GFAP, IL-1β, pCREB and pNF-κB proteins. The inhibition of AM activity did not change TNF-α-induced phosphorylation of extracellular signal-related kinase (pERK) while the treatment with AM1-50 still increased the level of pERK in the cultured DRG. Immunofluorescence assay showed the colocalization of AM-like immunoreactivity (IR) with TNF-α-IR in DRG neurons. The present study suggests that the increased AM receptor signaling mediated the many, but not all, TNF-α-induced activities, contributing to peripheral sensitization in neuropathic pain.
Collapse
Affiliation(s)
- Yajuan Chen
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350117, China
| | - Yan Zhang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350117, China
| | - Yuanhui Huo
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350117, China
| | - Dongmei Wang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350117, China
| | - Yanguo Hong
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University Fuzhou, Fujian 350117, China.
| |
Collapse
|
313
|
Transcriptional expression of inflammatory mediators in various somatosensory relay centers in the brain of rat models of peripheral mononeuropathy and local inflammation. J Neuroimmunol 2016; 297:81-91. [PMID: 27397080 DOI: 10.1016/j.jneuroim.2016.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/19/2016] [Accepted: 05/06/2016] [Indexed: 12/13/2022]
Abstract
Contradictory results have been reported regarding the role of inflammatory mediators in the central nervous system in mediating neuropathic pain and inflammatory hyperalgesia following peripheral nerve injury or localized inflammation. The present study aims to correlate between the mRNA expression and protein secretion of proinflammatory cytokines and nerve growth factor (NGF), in the dorsal root ganglia (DRGs), spinal cord, brainstem and thalamus, and pain-related behavior in animal models of peripheral mononeuropathy and localized inflammation. Different groups of rats (n=8, each) were subjected to either lesion of the nerves of their hindpaws to induce mononeuropathy or intraplantar injection of endotoxin (ET) and were sacrificed at various time intervals. TNF-α, IL-1β and NGF mRNA expression and protein levels in the various centers involved in processing nociceptive information were determined, by RT-PCR and ELISA. Control groups were either subjected to sham surgery or to saline injection. Mononeuropathy and ET injection produced significant and sustained increases in the mRNA expression and protein levels of TNF-α, IL-1β and NGF in the ipsilateral and contralateral DRGs, spinal cord, and brainstem. No significant and consistent changes in the mRNA expression of cytokines were noticed in the thalamus, while a downregulation of the NGF-mRNA level was observed. The temporal and spatial patterns of the observed changes in mRNA expression of cytokines and NGF are not closely in phase with the observed allodynia and hyperalgesia in the different models, suggesting that the role of these mediators may not be reduced exclusively to the production and maintenance of pain.
Collapse
|
314
|
George E, Elman I, Becerra L, Berg S, Borsook D. Pain in an era of armed conflicts: Prevention and treatment for warfighters and civilian casualties. Prog Neurobiol 2016; 141:25-44. [PMID: 27084355 DOI: 10.1016/j.pneurobio.2016.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/23/2016] [Accepted: 04/08/2016] [Indexed: 12/13/2022]
Abstract
Chronic pain is a common squealae of military- and terror-related injuries. While its pathophysiology has not yet been fully elucidated, it may be potentially related to premorbid neuropsychobiological status, as well as to the type of injury and to the neural alterations that it may evoke. Accordingly, optimized approaches for wounded individuals should integrate primary, secondary and tertiary prevention in the form of thorough evaluation of risk factors along with specific interventions to contravene and mitigate the ensuing chronicity. Thus, Premorbid Events phase may encompass assessments of psychological and neurobiological vulnerability factors in conjunction with fostering preparedness and resilience in both military and civilian populations at risk. Injuries per se phase calls for immediate treatment of acute pain in the field by pharmacological agents that spare and even enhance coping and adaptive capabilities. The key objective of the Post Injury Events is to prevent and/or reverse maladaptive peripheral- and central neural system's processes that mediate transformation of acute to chronic pain and to incorporate timely interventions for concomitant mental health problems including post-traumatic stress disorder and addiction We suggest that the proposed continuum of care may avert more disability and suffering than the currently employed less integrated strategies. While the requirements of the armed forces present a pressing need for this integrated continuum and a framework in which it can be most readily implemented, this approach may be also instrumental for the care of civilian casualties.
Collapse
Affiliation(s)
- E George
- Center for Pain and the Brain, Harvard Medical School (HMS), United States; Department of Anesthesia, Critical Care and Pain Medicine, MGH, HMS, Boston, MA, United States; Commander, MC, USN (Ret), United States
| | - I Elman
- Center for Pain and the Brain, Harvard Medical School (HMS), United States; Department of Psychiatry, Boonshoft School of Medicine and Dayton VA Medical Center, United States; Veterans Administration Medical Center, Dayton, OH, United States
| | - L Becerra
- Center for Pain and the Brain, Harvard Medical School (HMS), United States; Department of Anesthesia, Critical Care and Pain Medicine, BCH, HMS, Boston, MA, United States; Departments of Psychiatry and Radiology, MGH, Boston, MA, United States
| | - Sheri Berg
- Center for Pain and the Brain, Harvard Medical School (HMS), United States; Department of Anesthesia, Critical Care and Pain Medicine, MGH, HMS, Boston, MA, United States
| | - D Borsook
- Center for Pain and the Brain, Harvard Medical School (HMS), United States; Department of Anesthesia, Critical Care and Pain Medicine, BCH, HMS, Boston, MA, United States; Departments of Psychiatry and Radiology, MGH, Boston, MA, United States.
| |
Collapse
|
315
|
Abstract
Pathological pain is one of the most common neurological complications in patients with HIV-1/AIDS. However, the pathogenic process is unclear. Our recent studies show that Wnt5a is upregulated in the spinal cord dorsal horn (SDH) of the patients with HIV who develop pain and that HIV-1 gp120, a potential causal factor of the HIV-associated pain, rapidly upregulates Wnt5a in the mouse SDH. Using a mouse model, we show here that a specific Wnt5a antagonist, Box-5, attenuated gp120-induced mechanical allodynia. Conversely, a Wnt5a agonist, Foxy5, facilitated the allodynia. To elucidate the molecular mechanism by which Wnt5a regulates gp120-induced allodynia, we tested the role of the JNK/TNF-α pathway. We observed that the JNK-specific inhibitor SP600125 blocked either gp120- or Foxy5-induced allodynia. Similarly, the TNF-α-specific antagonist Enbrel also reversed either gp120- or Foxy5-induced allodynia. These data suggest that JNK and TNF-α mediate the biological effects of Wnt5a in regulating gp120-induced allodynia. To investigate the cellular mechanism, we performed extracellular single-unit recording from SDH neurons in anesthetized mice. Both Box-5 and SP600125 negated gp120-induced potentiation of SDH neuron spiking evoked by mechanical stimulation of the hind paw. Furthermore, while Foxy5 potentiated spike frequency of SDH neurons, either SP600125 or Enbrel blocked the potentiation. The data indicate that Wnt5a potentiates the activity of SDH neurons through the JNK-TNF-α pathway. Collectively, our findings suggest that Wnt5a regulates the pathogenesis of gp120-induced pain, likely by sensitizing pain-processing SDH neurons through JNK/TNF-α signaling.
Collapse
|
316
|
Galicia JC, Henson BR, Parker JS, Khan AA. Gene expression profile of pulpitis. Genes Immun 2016; 17:239-43. [PMID: 27052691 PMCID: PMC4892973 DOI: 10.1038/gene.2016.14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/27/2016] [Accepted: 02/03/2016] [Indexed: 01/27/2023]
Abstract
The cost, prevalence and pain associated with endodontic disease necessitate an understanding of the fundamental molecular aspects of its pathogenesis. This study was aimed to identify the genetic contributors to pulpal pain and inflammation. Inflamed pulps were collected from patients diagnosed with irreversible pulpitis (n=20). Normal pulps from teeth extracted for various reasons served as controls (n=20). Pain level was assessed using a visual analog scale (VAS). Genome-wide microarray analysis was performed using Affymetrix GeneTitan Multichannel Instrument. The difference in gene expression levels were determined by the Significance Analysis of Microarray program using a false discovery rate (q-value) of 5%. Genes involved in immune response, cytokine-cytokine receptor interaction and signaling, integrin cell surface interactions, and others were expressed at relatively higher levels in the in the pulpitis group. Moreover, several genes known to modulate pain and inflammation showed differential expression in asymptomatic and mild pain patients (≥30mm on VAS) compared to those with moderate to severe pain. This exploratory study provides a molecular basis for the clinical diagnosis of pulpitis. With an enhanced understanding of pulpal inflammation, future studies on treatment and management of pulpitis and on pain associated with it can have a biological reference to bridge treatment strategies with pulpal biology.
Collapse
Affiliation(s)
- J C Galicia
- Department of Endodontics, Arthur A Dugoni School of Dentistry, University of the Pacific, San Francisco, CA, USA.,Center for Pain Research and Innovation, Department of Endodontics, University of North Carolina School of Dentistry, Chapel Hill, NC, USA
| | - B R Henson
- DDS Program, University of North Carolina School of Dentistry, Chapel Hill, NC, USA
| | - J S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - A A Khan
- Center for Pain Research and Innovation, Department of Endodontics, University of North Carolina School of Dentistry, Chapel Hill, NC, USA
| |
Collapse
|
317
|
Kremer M, Yalcin I, Nexon L, Wurtz X, Ceredig RA, Daniel D, Hawkes RA, Salvat E, Barrot M. The antiallodynic action of pregabalin in neuropathic pain is independent from the opioid system. Mol Pain 2016; 12:12/0/1744806916633477. [PMID: 27030724 PMCID: PMC4956392 DOI: 10.1177/1744806916633477] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 01/22/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Clinical management of neuropathic pain, which is pain arising as a consequence of a lesion or a disease affecting the somatosensory system, partly relies on the use of anticonvulsant drugs such as gabapentinoids. Therapeutic action of gabapentinoids such as gabapentin and pregabalin, which act by the inhibition of calcium currents through interaction with the α2δ-1 subunit of voltage-dependent calcium channels, is well documented. However, some aspects of the downstream mechanisms are still to be uncovered. Using behavioral, genetic, and pharmacological approaches, we tested whether opioid receptors are necessary for the antiallodynic action of acute and/or long-term pregabalin treatment in the specific context of neuropathic pain. RESULTS Using the cuff model of neuropathic pain in mice, we show that acute pregabalin administration at high dose has a transitory antiallodynic action, while prolonged oral pregabalin treatment leads to sustained antiallodynic action, consistent with clinical observations. We show that pregabalin remains fully effective in μ-opioid receptor, in δ-opioid receptor and in κ-opioid receptor deficient mice, either female or male, and its antiallodynic action is not affected by acute naloxone. Our work also shows that long-term pregabalin treatment suppresses tumor necrosis factor-α overproduction induced by sciatic nerve constriction in the lumbar dorsal root ganglia. CONCLUSIONS We demonstrate that neither acute nor long-term antiallodynic effect of pregabalin in a context of neuropathic pain is mediated by the endogenous opioid system, which differs from opioid treatment of pain and antidepressant treatment of neuropathic pain. Our data are also supportive of an impact of gabapentinoid treatment on the neuroimmune aspect of neuropathic pain.
Collapse
Affiliation(s)
- Mélanie Kremer
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France Université de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Laurent Nexon
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Xavier Wurtz
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France Université de Strasbourg, Strasbourg, France
| | - Rhian Alice Ceredig
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France Université de Strasbourg, Strasbourg, France
| | - Dorothée Daniel
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Rachael Aredhel Hawkes
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France Université de Strasbourg, Strasbourg, France
| | - Eric Salvat
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France Centre d'Etude et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Michel Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| |
Collapse
|
318
|
Miranda HF, Noriega V, Prieto JC, Zanetta P, Castillo R, Aranda N, Sierralta F. Antinociceptive Interaction of Tramadol with Gabapentin in Experimental Mononeuropathic Pain. Basic Clin Pharmacol Toxicol 2016; 119:210-4. [DOI: 10.1111/bcpt.12567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/05/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Hugo F. Miranda
- Faculty of Medicine; School of Pharmacy; Andres Bello University; Santiago Chile
- Pharmacology Program; ICBM; Faculty Santiago; Chile of Medicine; University of Chile; Santiago 7 Chile
| | - Viviana Noriega
- Faculty of Medicine; School of Pharmacy; Andres Bello University; Santiago Chile
- Cardiovascular Department; Clinic Hospital; University of Chile; Santiago Chile
| | - Juan Carlos Prieto
- Pharmacology Program; ICBM; Faculty Santiago; Chile of Medicine; University of Chile; Santiago 7 Chile
- Cardiovascular Department; Clinic Hospital; University of Chile; Santiago Chile
| | - Pilar Zanetta
- Pharmacology Program; ICBM; Faculty Santiago; Chile of Medicine; University of Chile; Santiago 7 Chile
| | - Rodrigo Castillo
- Physiophathology Program; ICBM; Faculty of Medicine; University of Chile; Santiago 7 Chile
| | - Nicolás Aranda
- Pharmacology Program; ICBM; Faculty Santiago; Chile of Medicine; University of Chile; Santiago 7 Chile
| | - Fernando Sierralta
- Pharmacology Program; ICBM; Faculty Santiago; Chile of Medicine; University of Chile; Santiago 7 Chile
- Faculty of Odontology; Universidad Finis Terrae; Providencia Chile
| |
Collapse
|
319
|
Fong SW, Lin HC, Wu MF, Chen CC, Huang YS. CPEB3 Deficiency Elevates TRPV1 Expression in Dorsal Root Ganglia Neurons to Potentiate Thermosensation. PLoS One 2016; 11:e0148491. [PMID: 26915043 PMCID: PMC4767414 DOI: 10.1371/journal.pone.0148491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/19/2016] [Indexed: 01/07/2023] Open
Abstract
Cytoplasmic polyadenylation element binding protein 3 (CPEB3) is a sequence-specific RNA-binding protein that downregulates translation of multiple plasticity-related proteins (PRPs) at the glutamatergic synapses. Activity-induced synthesis of PRPs maintains long-lasting synaptic changes that are critical for memory consolidation and chronic pain manifestation. CPEB3-knockout (KO) mice show aberrant hippocampus-related plasticity and memory, so we investigated whether CPEB3 might have a role in nociception-associated plasticity. CPEB3 is widely expressed in the brain and peripheral afferent sensory neurons. CPEB3-KO mice with normal mechanosensation showed hypersensitivity to noxious heat. In the complete Freund's adjuvant (CFA)-induced inflammatory pain model, CPEB3-KO animals showed normal thermal hyperalgesia and transiently enhanced mechanical hyperalgesia. Translation of transient receptor potential vanilloid 1 (TRPV1) RNA was suppressed by CPEB3 in dorsal root ganglia (DRG), whereas CFA-induced inflammation reversed this inhibition. Moreover, CPEB3/TRPV1 double-KO mice behaved like TRPV1-KO mice, with severely impaired thermosensation and thermal hyperalgesia. An enhanced thermal response was recapitulated in non-inflamed but not inflamed conditional-KO mice, with cpeb3 gene ablated mostly but not completely, in small-diameter nociceptive DRG neurons. CPEB3-regulated translation of TRPV1 RNA may play a role in fine-tuning thermal sensitivity of nociceptors.
Collapse
Affiliation(s)
- Sitt Wai Fong
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsiu-Chen Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Meng-Fang Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
320
|
Wang X, Zhang Y, Peng Y, Hutchinson MR, Rice KC, Yin H, Watkins LR. Pharmacological characterization of the opioid inactive isomers (+)-naltrexone and (+)-naloxone as antagonists of toll-like receptor 4. Br J Pharmacol 2016; 173:856-69. [PMID: 26603732 DOI: 10.1111/bph.13394] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The toll-like receptor TLR4 is involved in neuropathic pain and in drug reward and reinforcement. The opioid inactive isomers (+)-naltrexone and (+)-naloxone act as TLR4 antagonists, reversing neuropathic pain and reducing opioid and cocaine reward and reinforcement. However, how these agents modulate TLR4 signalling is not clear. Here, we have elucidated the molecular mechanism of (+)-naltrexone and (+)-naloxone on TLR4 signalling. EXPERIMENTAL APPROACH BV-2 mouse microglial cell line, primary rat microglia and primary rat peritoneal macrophages were treated with LPS and TLR4 signalling inhibitors. Effects were measured using Western blotting, luciferase reporter assays, fluorescence microscopy and ELISA KEY RESULTS: (+)-Naltrexone and (+)-naloxone were equi-potent inhibitors of the LPS-induced TLR4 downstream signalling and induction of the pro-inflammatory factors NO and TNF-α. Similarly, (+)-naltrexone or (+)-naloxone inhibited production of reactive oxygen species and increased microglial phagocytosis, induced by LPS. However, (+)-naltrexone and (+)-naloxone did not directly inhibit the increased production of IL-1β, induced by LPS. The drug interaction of (+)-naloxone and (+)-naltrexone was additive. (+)-Naltrexone or (+)-naloxone inhibited LPS-induced activation of IFN regulatory factor 3 and production of IFN-β. However, they did not inhibit TLR4 signalling via the activation of either NF-κB, p38 or JNK in these cellular models. CONCLUSIONS AND IMPLICATIONS (+)-Naltrexone and (+)-naloxone were TRIF-IFN regulatory factor 3 axis-biased TLR4 antagonists. They blocked TLR4 downstream signalling leading to NO, TNF-α and reactive oxygen species. This pattern may explain, at least in part, the in vivo therapeutic effects of (+)-naltrexone and (+)-naloxone.
Collapse
Affiliation(s)
- X Wang
- Chemical Biology Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Y Zhang
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - Y Peng
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - M R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - K C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 20892, USA
| | - H Yin
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - L R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado at Boulder, Boulder, CO, 80309, USA
| |
Collapse
|
321
|
Bäckryd E, Ghafouri B, Larsson B, Gerdle B. Plasma pro-inflammatory markers in chronic neuropathic pain: A multivariate, comparative, cross-sectional pilot study. Scand J Pain 2016; 10:1-5. [PMID: 28361755 DOI: 10.1016/j.sjpain.2015.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/24/2015] [Accepted: 06/26/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND Caused by a lesion or disease of the somatosensory system, neuropathic pain is notoriously difficult to treat with conventional analgesics. It has been suggested that inflammatory cytokines play a role in the development and maintenance of neuropathic pain. But human studies of these substances are relatively few and partly contradictory. OBJECTIVES To simultaneously investigate the plasma levels of chemokine interleukin 8 (IL-8) and the cytokines IL-6, IL-1β, and Granulocyte macrophage colony-stimulating factor (GM-CSF) in patients with peripheral neuropathic pain (most of whom due to failed back surgery syndrome) (n=14) compared to controls (n=17). RESULTS IL-6 was significantly higher in patients than in controls (0.92±0.12pg/ml vs. 0.57±0.08pg/ml, p=0.012). IL-1β, IL-8, and GM-CSF levels did not differ between the two groups. A multivariate analysis showed a tendency for patients also to have higher GM-CSF plasma levels than controls. CONCLUSIONS This study found an increased level of IL-6 in plasma in patients with neuropathic pain, but not for the other pro-inflammatory substances investigated. There are several possible confounders not registered or controlled for in this and other studies of neuropathic pain. IMPLICATIONS Larger studies that take several possible confounders into consideration are needed to further investigate the levels of plasma cytokines in different pain conditions.
Collapse
Affiliation(s)
- Emmanuel Bäckryd
- Division of Community Medicine, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University,LinköpingSweden
- Pain and Rehabilitation Centre, Anaesthetics, Operations and Specialty Surgery Centre, Region Östergötland, Sweden
| | - Bijar Ghafouri
- Division of Community Medicine, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University,LinköpingSweden
- Pain and Rehabilitation Centre, Anaesthetics, Operations and Specialty Surgery Centre, Region Östergötland, Sweden
| | - Britt Larsson
- Division of Community Medicine, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University,LinköpingSweden
- Pain and Rehabilitation Centre, Anaesthetics, Operations and Specialty Surgery Centre, Region Östergötland, Sweden
| | - Björn Gerdle
- Division of Community Medicine, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University,LinköpingSweden
- Pain and Rehabilitation Centre, Anaesthetics, Operations and Specialty Surgery Centre, Region Östergötland, Sweden
| |
Collapse
|
322
|
Wang J, Ding CP, Yu J, Zeng XY, Han SP, Wang JY. Dynamic distributions of tumor necrosis factor-alpha and its receptors in the red nucleus of rats with spared nerve injury. Neuropathology 2015; 36:346-53. [PMID: 26669937 DOI: 10.1111/neup.12282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/12/2023]
Abstract
Previous studies have demonstrated that tumor necrosis factor-alpha (TNF-α) in the red nucleus (RN) plays a facilitated role in the development of neuropathic pain, and its effect is transmitted through TNF-α receptor (TNFR) subtypes 1 and 2. Here, the dynamic distributions of TNF-α and TNFRs in the RN of rats with spared nerve injury (SNI) were investigated. Western blot analysis and immunofluorescence staining indicated that TNF-α was hardly expressed in the RN of normal rats but significantly increased at 1 week and peaked at 2 weeks after SNI. Neurons and oligodendrocytes showed TNF-α expression at both 1 week and 2 weeks after SNI, while astrocytes and microglia produced TNF-α later than neurons and oligodendrocytes starting at 2 weeks after SNI. TNFR1 was constitutively expressed in the RN of normal rats and significantly enhanced at 2 weeks but not 1 week after SNI; it was mainly localized in neurons, oligodendrocytes and microglia. Astrocytes were not immunopositive for TNFR1 under normal conditions and at 1 week after injury, but small amounts of astrocytes showed TNFR1 expression at 2 weeks after SNI. A low level of TNFR2 was expressed in the RN of normal rats, but it was significantly increased at 1 week and 2 weeks after SNI and localized in neurons and all three types of glia. These findings suggest that neurons and three types of glia in the RN all contribute to TNF-α production and participate in the initiation and/or maintenance of neuropathic pain induced by SNI. TNF-α exerts its effects in different types of cells maybe through different receptors, TNFR1 and/or TNFR2, in the different stages of neuropathic pain.
Collapse
Affiliation(s)
- Jing Wang
- Departments of Pathogenic Biology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Cui-Ping Ding
- Departments of Pathogenic Biology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jing Yu
- Departments of Pathogenic Biology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xiao-Yan Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Shui-Ping Han
- Department of Pathology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jun-Yang Wang
- Departments of Pathogenic Biology and Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
323
|
Chen Y, Li G, Huang LYM. p38 MAPK mediates glial P2X7R-neuronal P2Y1R inhibitory control of P2X3R expression in dorsal root ganglion neurons. Mol Pain 2015; 11:68. [PMID: 26542462 PMCID: PMC4635984 DOI: 10.1186/s12990-015-0073-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
Background We have previously shown that endogenously active purinergic P2X7 receptors (P2X7Rs) in satellite glial cells of dorsal root ganglia (DRGs) stimulate ATP release. The ATP activates P2Y1Rs located in the enwrapped neuronal somata, resulting in down-regulation of P2X3Rs. This P2X7R-P2Y1-P2X3R inhibitory control significantly reduces P2X3R-mediated nociceptive responses. The underlying mechanism by which the activation of P2Y1Rs inhibits the expression of P2X3Rs remains unexplored. Results Examining the effect of the activation of p38 mitogen-activated protein kinase on the expression of P2X3Rs in DRGs, we found that the p38 activator, anisomycin (Anis), reduced the expression of P2X3Rs. Blocking the activity of SGCs by the glial Krebs cycle inhibitor, fluorocitrate, did not change the effect of Anis. These results suggest that neuronal p38 plays a major role in the inhibition of P2X3R expression. Western blotting analyses showed that inhibiting P2Y1Rs by MRS2179 (MRS) or blocking P2X7Rs by either oxATP or A740003 reduced pp38 and increased P2X3R expression in DRGs. These results are further supported by the immunohistochemical study showing that P2X7R and P2Y1R antagonists reduce the percentage of pp38-positive neurons. These observations suggest that activation of P2X7Rs and P2Y1Rs promotes p38 activity to exert inhibitory control on P2X3R expression. Since activation of p38 by Anis in the presence of either A740003 or MRS could overcome the block of P2X7R-P2Y1R inhibitory control, p38 in DRG neurons is downstream of P2Y1Rs. In addition, inhibition of p38 by SB202190 was found to prevent the P2X7R and P2Y1R block of P2X3R expression and increase P2X3R-mediated nociceptive flinch behaviors. Conclusions p38 in DRG neurons downstream of P2Y1R is necessary and sufficient for the P2X7R-P2Y1R inhibitory control of P2X3R expression.
Collapse
Affiliation(s)
- Yong Chen
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555-1069, USA.
| | - Guangwen Li
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555-1069, USA.
| | - Li-Yen Mae Huang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555-1069, USA.
| |
Collapse
|
324
|
Laedermann CJ, Abriel H, Decosterd I. Post-translational modifications of voltage-gated sodium channels in chronic pain syndromes. Front Pharmacol 2015; 6:263. [PMID: 26594175 PMCID: PMC4633509 DOI: 10.3389/fphar.2015.00263] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/23/2015] [Indexed: 02/06/2023] Open
Abstract
In the peripheral sensory nervous system the neuronal expression of voltage-gated sodium channels (Navs) is very important for the transmission of nociceptive information since they give rise to the upstroke of the action potential (AP). Navs are composed of nine different isoforms with distinct biophysical properties. Studying the mutations associated with the increase or absence of pain sensitivity in humans, as well as other expression studies, have highlighted Nav1.7, Nav1.8, and Nav1.9 as being the most important contributors to the control of nociceptive neuronal electrogenesis. Modulating their expression and/or function can impact the shape of the AP and consequently modify nociceptive transmission, a process that is observed in persistent pain conditions. Post-translational modification (PTM) of Navs is a well-known process that modifies their expression and function. In chronic pain syndromes, the release of inflammatory molecules into the direct environment of dorsal root ganglia (DRG) sensory neurons leads to an abnormal activation of enzymes that induce Navs PTM. The addition of small molecules, i.e., peptides, phosphoryl groups, ubiquitin moieties and/or carbohydrates, can modify the function of Navs in two different ways: via direct physical interference with Nav gating, or via the control of Nav trafficking. Both mechanisms have a profound impact on neuronal excitability. In this review we will discuss the role of Protein Kinase A, B, and C, Mitogen Activated Protein Kinases and Ca++/Calmodulin-dependent Kinase II in peripheral chronic pain syndromes. We will also discuss more recent findings that the ubiquitination of Nav1.7 by Nedd4-2 and the effect of methylglyoxal on Nav1.8 are also implicated in the development of experimental neuropathic pain. We will address the potential roles of other PTMs in chronic pain and highlight the need for further investigation of PTMs of Navs in order to develop new pharmacological tools to alleviate pain.
Collapse
Affiliation(s)
- Cedric J. Laedermann
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Harvard Medical School, BostonMA, USA
| | - Hugues Abriel
- Department of Clinical Research, University of BernBern, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of LausanneLausanne, Switzerland
- Department of Fundamental Neurosciences, University of LausanneLausanne, Switzerland
| |
Collapse
|
325
|
Lee JB, Byun JH, Choi IS, Kim Y, Lee JS. The Effect of Pulsed Radiofrequency Applied to the Peripheral Nerve in Chronic Constriction Injury Rat Model. Ann Rehabil Med 2015; 39:667-75. [PMID: 26605164 PMCID: PMC4654073 DOI: 10.5535/arm.2015.39.5.667] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/29/2015] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To investigate the effect of pulsed radiofrequency (PRF) applied proximal to the injured peripheral nerve on the expression of tumor necrosis factor-α (TNF-α) in a neuropathic pain rat model. METHODS Nineteen male Sprague-Dawley rats were used in the study. All rats underwent chronic constriction injury (CCI) procedure. After 7 days of CCI, withdrawal frequency of affected hind paw to mechanical stimuli and withdrawal latency of affected hind paw to heat stimulus were measured. They were randomly divided into two groups: group A, CCI group (n=9) and group B, CCI treated with PRF group (n=10). Rats of group B underwent PRF procedure on the sciatic nerve. Withdrawal frequency and withdrawal latency were measured at 12 hours, and 7 days after PRF. Immunohistochemistry and Western blot analysis were performed using a TNF-α antibody. RESULTS Before PRF, withdrawal frequency and withdrawal latency were not different in both groups. After PRF, withdrawal frequency decreased and withdrawal latency prolonged over time in group B. There was significant interaction between time and group for each withdrawal frequency and withdrawal latency. Group B showed decreased TNF-α immunoreactivity of the spinal cord and sciatic nerve at 7 days. CONCLUSION PRF applied proximal to the peripheral nerve injury is potentially helpful for the reduction of neuropathic pain by neuromodulation of inflammatory markers.
Collapse
Affiliation(s)
- Jun-Beom Lee
- Department of Physical and Rehabilitation Medicine, Research Institute of Medical Sciences, Chonnam National University Medical School & Hospital, Gwangju, Korea
| | - Jeong-Hyun Byun
- Department of Physical and Rehabilitation Medicine, Research Institute of Medical Sciences, Chonnam National University Medical School & Hospital, Gwangju, Korea
| | - In-Sung Choi
- Department of Physical and Rehabilitation Medicine, Research Institute of Medical Sciences, Chonnam National University Medical School & Hospital, Gwangju, Korea
| | - Young Kim
- Department of Pathology, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea
| | - Ji Shin Lee
- Department of Pathology, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
326
|
Tumor necrosis factor inhibitor therapy in ankylosing spondylitis: differential effects on pain and fatigue and brain correlates. Pain 2015; 156:297-304. [PMID: 25599451 DOI: 10.1097/01.j.pain.0000460310.71572.16] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Ankylosing spondylitis is associated with back pain and fatigue and impacts mobility but can be treated with tumor necrosis factor inhibitors (TNFi). The differential effects of TNFi treatment on multiple symptoms and the brain is not well delineated. Thus, we conducted a 2-part study. In study 1, we conducted a retrospective chart review in 129 ankylosing spondylitis patients to assess TNFi effects on pain, fatigue, motor function, mobility, and quality of life (QoL). After at least 10 weeks of TNFi treatment, patients had clinically significant improvements (>30%) in pain (including neuropathic pain), most disease and QoL factors, and normalized sensory detection thresholds. However, residual fatigue (mean = 5.3) was prominent. Although 60% of patients had significant relief of pain, only 22% of patients had significant relief of both pain and fatigue. Therefore, the preferential TNFi treatment effect on pain compared with fatigue could contribute to suboptimal effects on QoL. Part 2 was a prospective study in 14 patients to identify TNFi treatment effects on pain, fatigue, sensory and psychological factors, and brain cortical thickness based on 3T magnetic resonance imaging. Centrally, TNFi was associated with statistically significant cortical thinning of motor, premotor, and posterior parietal regions. Pain intensity reduction was associated with cortical thinning of the secondary somatosensory cortex, and pain unpleasantness reduction was associated with the cortical thinning of motor areas. In contrast, fatigue reduction correlated with cortical thinning of the insula, primary sensory cortex/inferior parietal sulcus, and superior temporal polysensory areas. This indicates that TNFi treatment produces changes in brain areas implicated in sensory, motor, affective, and cognitive functions.
Collapse
|
327
|
Li Q, Zhang X. Epigallocatechin-3-gallate attenuates bone cancer pain involving decreasing spinal Tumor Necrosis Factor-α expression in a mouse model. Int Immunopharmacol 2015; 29:818-823. [PMID: 26363974 DOI: 10.1016/j.intimp.2015.08.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/19/2015] [Accepted: 08/27/2015] [Indexed: 11/30/2022]
Abstract
Tumor metastasis to bone often elicits a wide array of symptoms, in which pain is a significant factor in catastrophic complications of bone cancer. The complete understanding of bone cancer-related pain is still unknown, while several pathophysiological components have been suggested, from tumor-stimulated osteolysis, nerve compression, stimulations of ion channels, and locally generated inflammatory cytokines. In particular, it has been shown that pro-inflammatory cytokine TNFα-mediated actions are necessary for the development of bone cancer pain. As a member of catechin family in green tea extracts, EGCG (Epigallocatechin-3-gallate) can reduce excess free radicals and attenuate overactive inflammatory signaling including TNFα. In addition, EGCG or its related molecules have been used to control neuropathic pain in various preclinical settings. However, its potential use in bone cancer-caused pain has not yet been reported. Here we show that treating a mouse model of bone cancer by EGCG, results in a dramatic reduction in pain behavior and a significant decrease of TNFα expression within the spinal cord of tumor-bearing mice. Thus, this study reveals an anti-nociceptive role for EGCG in the progression of pain caused by tumor bone metastasis, and highlights a potential scheme by using anti-TNFα as a therapeutic option for osteolytic pain.
Collapse
Affiliation(s)
- Qingsong Li
- Department of Anesthesiology, The Second Hospital of Shandong University, No. 247 Beiyuan Road, Jinan 250033, China.
| | - Xi Zhang
- Department of Anesthesiology, The Second Hospital of Shandong University, No. 247 Beiyuan Road, Jinan 250033, China
| |
Collapse
|
328
|
Wang C, Jiang Q, Wang M, Li D. MiR-19a targets suppressor of cytokine signaling 1 to modulate the progression of neuropathic pain. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:10901-10907. [PMID: 26617805 PMCID: PMC4637620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/21/2015] [Indexed: 06/05/2023]
Abstract
PURPOSE We aimed to investigate whether miR-19a is associated with neuropathic pain and elucidate the underlying regulatory mechanism. METHODS We established a neuropathic pain model of bilateral chronic constriction injury (bCCI). Then bCCI rats were injected with mo-miR-19a, siR-SOCS1 or blank expression vector through a microinjection syringe via an intrathecal catheter on 3 day before surgery and after surgery. Behavioral tests, such as mechanical allodynia, thermal hyperalgesia and acetone induced cold allodynia, were performed to evaluate the pain threshold. Besides, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to determine the expression of miR-19a and western blotting was carried out to measure the expression of SOCS1. RESULTS miR-19a expression levels were markedly increased in neuropathic pain models. Moreover, miR-19a significantly attenuated mechanical allodynia and thermal hyperalgesia, and similar results were obtained after knockdown of SOCS1 expression. However, miR-19a markedly increased the times that the rats appeared a sign of cold allodynia, and knockdown of SOCS1 expression had similar effects. Besides, the results of bioinformatics analysis and western blotting analysis were all confirmed that SOCS1 was a direct target of miR-19a in neuropathic pain models. CONCLUSIONS Our finding indicate that SOCS1 is a direct target of miR-19a in neuropathic pain rats and miR-19a may play a critical role in regulating of neuropathic pain via targeting SOCS1.
Collapse
Affiliation(s)
- Conghui Wang
- Department of Anesthesiology, The First Hospital of Xiangyang City Xiangyang 441000, Hubei Province, China
| | - Qi Jiang
- Department of Anesthesiology, The First Hospital of Xiangyang City Xiangyang 441000, Hubei Province, China
| | - Min Wang
- Department of Anesthesiology, The First Hospital of Xiangyang City Xiangyang 441000, Hubei Province, China
| | - Dong Li
- Department of Anesthesiology, The First Hospital of Xiangyang City Xiangyang 441000, Hubei Province, China
| |
Collapse
|
329
|
Stemkowski PL, Noh MC, Chen Y, Smith PA. Increased excitability of medium-sized dorsal root ganglion neurons by prolonged interleukin-1β exposure is K(+) channel dependent and reversible. J Physiol 2015; 593:3739-55. [PMID: 26110238 PMCID: PMC4560594 DOI: 10.1113/jp270905] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/17/2015] [Indexed: 01/27/2023] Open
Abstract
KEY POINTS Neuropathic pain resulting from peripheral nerve injury is initiated and maintained by persistent ectopic activity in primary afferent neurons. Sciatic nerve injury increases the excitability of medium-sized dorsal root ganglion (DRG) neurons. Levels of the inflammatory cytokine interleukin 1β (IL-1β) increase and peak after 7 days. Five to six days of exposure of medium sized DRG neurons to 100 pm IL-1β promotes persistent increases in excitability which abate within 3-4 days of cytokine removal. This is associated with a profound attenuation of K(+) channel currents but only modest increases in function of cyclic nucleotide-sensitive hyperpolarization-activated channels (HCNs) and of voltage-gated Na(+) and Ca(2+) channel currents. It is unlikely, therefore, that direct interaction of IL-1β with DRG neurons is capable of initiating an enduring phenotypic shift in their electrophysiological properties that follows sciatic nerve injury. The findings also underline the importance of K(+) channel modulation in the actions of inflammatory mediators on peripheral neurons. ABSTRACT Chronic constriction injury of rat sciatic nerve promotes signs of neuropathic pain. This is associated with an increase in the level of interleukin 1β (IL-1β) in primary afferents that peaks at 7 days. This initial cytokine exposure has been proposed to trigger an enduring alteration in neuronal phenotype that underlies chronic hyper-excitability in sensory nerves, which initiates and maintains chronic neuropathic pain. We have shown previously that 5-6 days of exposure of rat dorsal root ganglia (DRGs) to 100 pm IL-1β increases the excitability of medium-sized neurons. We have now found using whole-cell recording that this increased excitability reverts to control levels within 3-4 days of cytokine removal. The effects of IL-1β were dominated by changes in K(+) currents. Thus, the amplitudes of A-current, delayed rectifier and Ca(2+) -sensitive K(+) currents were reduced by ∼68%, ∼64% and ∼36%, respectively. Effects of IL-1β on other cation currents were modest by comparison. There was thus a slight decrease in availability of high voltage-activated Ca(2+) channel current, a small increase in rates of activation of hyperpolarization-activated cyclic nucleotide-gated channel current (IH ), and a shift in the voltage dependence of activation of tetrodotoxin-sensitive sodium current (TTX-S INa ) to more negative potentials. It is unlikely, therefore, that direct interaction of IL-1β with DRG neurons initiates an enduring phenotypic shift in their electrophysiological properties following sciatic nerve injury. Persistent increases in primary afferent excitability following nerve injury may instead depend on altered K(+) channel function and on the continued presence of slightly elevated levels IL-1β and other cytokines.
Collapse
Affiliation(s)
- Patrick L Stemkowski
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada, T2N 4N1
| | - Myung-Chul Noh
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Yishen Chen
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Peter A Smith
- Centre for Neuroscience and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| |
Collapse
|
330
|
Papanas N, Ziegler D. Risk Factors and Comorbidities in Diabetic Neuropathy: An Update 2015. Rev Diabet Stud 2015; 12:48-62. [PMID: 26676661 PMCID: PMC5397983 DOI: 10.1900/rds.2015.12.48] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 04/27/2015] [Accepted: 05/07/2015] [Indexed: 01/01/2023] Open
Abstract
Distal symmetric sensorimotor polyneuropathy (DSPN) is the most common neurological manifestation in diabetes. Major risk factors of DSPN include diabetes duration, hyperglycemia, and age, followed by prediabetes, hypertension, dyslipidemia, and obesity. Height, smoking, insulin resistance, hypoinsulinemia, and others represent an additional risk. Importantly, hyperglycemia, hypertension, dyslipidemia, obesity, and smoking are modifiable. Stringent glycemic control has been shown to be effective in type 1, but not to the same extent in type 2 diabetes. Antilipidemic treatment, especially with fenofibrate, and multi-factorial intervention have produced encouraging results, but more experience is necessary. The major comorbidities of DSPN are depression, autonomic neuropathy, peripheral arterial disease, cardiovascular disease, nephropathy, retinopathy, and medial arterial calcification. Knowledge of risk factors and comorbidities has the potential to enrich the therapeutic strategy in clinical practice as part of the overall medical care for patients with neuropathy. This article provides an updated overview of DSPN risk factors and comorbidities.
Collapse
Affiliation(s)
- Nikolaos Papanas
- Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
331
|
Pillarisetti S, Khanna I. A multimodal disease modifying approach to treat neuropathic pain--inhibition of soluble epoxide hydrolase (sEH). Drug Discov Today 2015; 20:1382-90. [PMID: 26259523 DOI: 10.1016/j.drudis.2015.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/23/2015] [Accepted: 07/30/2015] [Indexed: 01/05/2023]
Abstract
Both neuronal and non-neuronal mechanisms have been proposed to contribute to neuropathic pain (NP). All currently approved treatments for NP modulate neuronal targets and provide only symptomatic relief. Here we review evidence that inhibition of soluble epoxide hydrolase (sEH), the enzyme that degrades epoxyeicosatrienoic acids (EETs), has potential to be a multimodal, disease modifying approach to treat NP: (1) EET actions involve both endogenous opioid system and the GABAergic systems thus provide superior pain relief compared to morphine or gabapentin, (2) EETs are directly anti-inflammatory and inhibit expression of inflammatory cytokines and adhesion molecules thus can prevent continued nerve damage; and (3) EETs promote nerve regeneration in cultured neurons. Thus, an sEH inhibitor will not only provide effective pain relief, but would also block further nerve damage and promote healing.
Collapse
|
332
|
Pandurangan K, Krishnappan V, Subramanian V, Subramanyan R. Anti-inflammatory effect of certain dimethoxy flavones. Inflammopharmacology 2015; 23:307-17. [PMID: 26248971 DOI: 10.1007/s10787-015-0242-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/29/2015] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The aim of the present study was to evaluate the anti-inflammatory effect of four dimethoxy flavone derivatives; 7,2'-dimethoxy flavone, 7,3'-dimethoxy flavone, 7,4'-dimethoxy flavone and 7,8,-dimethoxy flavone and to investigate the possible cellular mechanisms involved. MATERIALS AND METHODS The acute anti-inflammatory effect of dimethoxy flavones was investigated by carrageenan induced hind paw oedema in rats. Further, the effect of dimethoxy flavones on certain mediators of pain and inflammation like cyclooxygenases (COX-1 and COX-2), pro-inflammatory cytokines (IL-1β and TNF-α) and free radical scavenging activity (NO and LPO) were investigated by using in vitro tests. RESULTS The investigated dimethoxy flavones produced a significant, dose and time dependent reduction of carrageenan induced paw oedema in rats with a maximum inhibition of 52.4% observed for 7,4'-dimethoxy flavone. Although, the test compounds inhibited both the isoforms of cyclooxygenase, a higher degree of inhibition on COX-2 was evident. A concentration dependent inhibition of other inflammatory cytokines like tumor necrosis factor-α and interleukin-1β was identified in the present study. 7,4'-dimethoxy flavone was found to be maximally effective in inhibiting nitrite ion free radical generation and 7,8-dimethoxy flavone was more active in inhibiting lipid peroxidation than the other compounds. CONCLUSION The results of the present study reveal the anti-inflammatory action of the investigated dimethoxy flavones. Inhibition of cyclooxygenases, cytokines and reactive oxygen species, observed in subsequent experiments may be suggested as possible mechanisms involved in the action of these compounds.
Collapse
Affiliation(s)
- Kamalakannan Pandurangan
- Department of Pharmacology, Meenakshi Medical College and Research Institute, Meenakshi Academy of Higher Education and Research, Kanchipuram, 631 552, India.
| | | | - Viswanathan Subramanian
- Department of Pharmacology, Meenakshi Medical College and Research Institute, Meenakshi Academy of Higher Education and Research, Kanchipuram, 631 552, India
| | - Ramaswamy Subramanyan
- Department of Pharmacology, Sri Lakshminarayana Institute of Medical Sciences, Pondicherry, 605 502, India
| |
Collapse
|
333
|
Galbavy W, Kaczocha M, Puopolo M, Liu L, Rebecchi MJ. Neuroimmune and Neuropathic Responses of Spinal Cord and Dorsal Root Ganglia in Middle Age. PLoS One 2015; 10:e0134394. [PMID: 26241743 PMCID: PMC4524632 DOI: 10.1371/journal.pone.0134394] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023] Open
Abstract
Prior studies of aging and neuropathic injury have focused on senescent animals compared to young adults, while changes in middle age, particularly in the dorsal root ganglia (DRG), have remained largely unexplored. 14 neuroimmune mRNA markers, previously associated with peripheral nerve injury, were measured in multiplex assays of lumbar spinal cord (LSC), and DRG from young and middle-aged (3, 17 month) naïve rats, or from rats subjected to chronic constriction injury (CCI) of the sciatic nerve (after 7 days), or from aged-matched sham controls. Results showed that CD2, CD3e, CD68, CD45, TNF-α, IL6, CCL2, ATF3 and TGFβ1 mRNA levels were substantially elevated in LSC from naïve middle-aged animals compared to young adults. Similarly, LSC samples from older sham animals showed increased levels of T-cell and microglial/macrophage markers. CCI induced further increases in CCL2, and IL6, and elevated ATF3 mRNA levels in LSC of young and middle-aged adults. Immunofluorescence images of dorsal horn microglia from middle-aged naïve or sham rats were typically hypertrophic with mostly thickened, de-ramified processes, similar to microglia following CCI. Unlike the spinal cord, marker expression profiles in naïve DRG were unchanged across age (except increased ATF3); whereas, levels of GFAP protein, localized to satellite glia, were highly elevated in middle age, but independent of nerve injury. Most neuroimmune markers were elevated in DRG following CCI in young adults, yet middle-aged animals showed little response to injury. No age-related changes in nociception (heat, cold, mechanical) were observed in naïve adults, or at days 3 or 7 post-CCI. The patterns of marker expression and microglial morphologies in healthy middle age are consistent with development of a para-inflammatory state involving microglial activation and T-cell marker elevation in the dorsal horn, and neuronal stress and satellite cell activation in the DRG. These changes, however, did not affect the establishment of neuropathic pain.
Collapse
Affiliation(s)
- William Galbavy
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Lixin Liu
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Mario J Rebecchi
- Department of Anesthesiology, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
334
|
Gerard E, Spengler RN, Bonoiu AC, Mahajan SD, Davidson BA, Ding H, Kumar R, Prasad PN, Knight PR, Ignatowski TA. Chronic constriction injury-induced nociception is relieved by nanomedicine-mediated decrease of rat hippocampal tumor necrosis factor. Pain 2015; 156:1320-1333. [PMID: 25851457 PMCID: PMC4474806 DOI: 10.1097/j.pain.0000000000000181] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuropathic pain is a chronic pain syndrome that arises from nerve injury. Current treatments only offer limited relief, clearly indicating the need for more effective therapeutic strategies. Previously, we demonstrated that proinflammatory tumor necrosis factor-alpha (TNF) is a key mediator of neuropathic pain pathogenesis; TNF is elevated at sites of neuronal injury, in the spinal cord, and supraspinally during the initial development of pain. The inhibition of TNF action along pain pathways outside higher brain centers results in transient decreases in pain perception. The objective of this study was to determine whether specific blockade of TNF in the hippocampus, a site of pain integration, could prove efficacious in reducing sciatic nerve chronic constriction injury (CCI)-induced pain behavior. Small inhibitory RNA directed against TNF mRNA was complexed to gold nanorods (GNR-TNF siRNA; TNF nanoplexes) and injected into the contralateral hippocampus of rats 4 days after unilateral CCI. Withdrawal latencies to a noxious thermal stimulus (hyperalgesia) and withdrawal to innocuous forces (allodynia) were recorded up to 10 days and compared with baseline values and sham-operated rats. Thermal hyperalgesia was dramatically decreased in CCI rats receiving hippocampal TNF nanoplexes; and mechanical allodynia was transiently relieved. TNF levels (bioactive protein, TNF immunoreactivity) in hippocampal tissue were decreased. The observation that TNF nanoplex injection into the hippocampus alleviated neuropathic pain-like behavior advances our previous findings that hippocampal TNF levels modulate pain perception. These data provide evidence that targeting TNF in the brain using nanoparticle-protected siRNA may be an effective strategy for treatment of neuropathic pain.
Collapse
Affiliation(s)
- Elizabeth Gerard
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York
| | | | - Adela C. Bonoiu
- Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York
| | - Supriya D. Mahajan
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, University at Buffalo, The State University of New York
| | - Bruce A. Davidson
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York
- NanoAxis, LLC, Clarence, New York 14031
- Department of Anesthesiology, University at Buffalo, The State University of New York
- Veterans Administration Western New York Healthcare System
| | - Hong Ding
- Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York
| | - Rajiv Kumar
- Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York
| | - Paras N. Prasad
- Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York
- Department of Chemistry, University at Buffalo, The State University of New York
| | - Paul R. Knight
- NanoAxis, LLC, Clarence, New York 14031
- Institute for Lasers, Photonics, and Biophotonics, University at Buffalo, The State University of New York
- Department of Anesthesiology, University at Buffalo, The State University of New York
- Veterans Administration Western New York Healthcare System
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York
| | - Tracey A. Ignatowski
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York
- NanoAxis, LLC, Clarence, New York 14031
- Program for Neuroscience, University at Buffalo, The State University of New York
| |
Collapse
|
335
|
DeConde AS, Mace JC, Ashby S, Smith TL, Orlandi RR, Alt JA. Characterization of facial pain associated with chronic rhinosinusitis using validated pain evaluation instruments. Int Forum Allergy Rhinol 2015; 5:682-90. [PMID: 26074476 DOI: 10.1002/alr.21539] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/11/2015] [Accepted: 03/05/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Prior investigations into facial pain associated with chronic rhinosinusitis (CRS) have yielded important results, but have yet to use pain-specific outcome measures. This study seeks to characterize facial pain associated with CRS using validated pain-specific instruments. METHODS Adults with CRS were enrolled into a prospective, cross-sectional study along with control participants presenting with non-CRS diagnoses. Facial pain was characterized in both groups using the Brief Pain Inventory Short Form (BPI-SF) and the Short-Form McGill Pain Questionnaire (SF-MPQ). CRS-specific measures of disease were measured including the 22-item Sino-Nasal Outcome Test-22 (SNOT-22), nasal endoscopy, and computed tomography scoring. RESULTS The patients comprised of CRS with nasal polyposis (CRSwNP; n = 25), CRS without nasal polyposis (CRSsNP; n = 30), and control participants (n = 8). Subjects with CRSwNP and CRSsNP were less likely to be pain free than controls (16.0%, 6.7%, and 62.5% respectively, p = 0.001) and carried greater burden of pain as measured by the BPI-SF and SF-MPQ than controls (p = 0.002 and p = 0.017, respectively). Pain in CRS was most commonly located around the eyes and characterized as "throbbing" and "aching." Nasal polyp status was not associated with differences in character, severity, or location of pain. CONCLUSION Subjects with CRS have a greater burden of facial pain relative to control subjects across several standardized pain measures. Further, facial pain in CRS significantly correlated to quality of life and CRS-specific disease severity measures. Study across larger cohorts using standardized pain measures is warranted to clarify the association of facial pain with CRS.
Collapse
Affiliation(s)
- Adam S DeConde
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of California San Diego, San Diego, CA
| | - Jess C Mace
- Department of Otolaryngology-Head and Neck Surgery, Division of Rhinology, Oregon Health and Science University, Portland, OR
| | - Shaelene Ashby
- Sinus and Skull Base Surgery Program, Division of Otolaryngology-Head and Neck Surgery, Department of Surgery; University of Utah, Salt Lake City, UT
| | - Timothy L Smith
- Department of Otolaryngology-Head and Neck Surgery, Division of Rhinology, Oregon Health and Science University, Portland, OR
| | - Richard R Orlandi
- Sinus and Skull Base Surgery Program, Division of Otolaryngology-Head and Neck Surgery, Department of Surgery; University of Utah, Salt Lake City, UT
| | - Jeremiah A Alt
- Sinus and Skull Base Surgery Program, Division of Otolaryngology-Head and Neck Surgery, Department of Surgery; University of Utah, Salt Lake City, UT
| |
Collapse
|
336
|
van der Wal S, Cornelissen L, Behet M, Vaneker M, Steegers M, Vissers K. Behavior of neuropathic pain in mice following chronic constriction injury comparing silk and catgut ligatures. SPRINGERPLUS 2015; 4:225. [PMID: 26069872 PMCID: PMC4456577 DOI: 10.1186/s40064-015-1009-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/29/2015] [Indexed: 01/01/2023]
Abstract
Introduction Neuropathic pain is defined as pain arising as a direct consequence of a lesion or disease affecting the somatosensory system and is common after surgery. Neuropathic pain can persist without an obvious injury. In this study we aim to validate a murine chronic constriction injury model as a model for neuropathic pain research and determine if silk or catgut ligatures induced most stable neuropathic pain behavior. Methods In this study mice underwent chronic constriction or sham surgery. Mice were tested on cutaneous hyperalgesia with the cumulative reaction time in the acetone test, on allodynia with the cumulative reaction time and number of lifts in the cold plate test and the maximal force before withdrawal in von Frey test. Results In the acetone test neuropathic pain was seen in CCI mice, but not in sham mice. Hyperalgesia was present postoperatively in CCI mice compared with preoperatively. In the cold plate test cumulative reaction time and number of lifts were higher in the ipsilateral hind paw than in the contralateral hind paw and sham mice. Postoperative measurements were higher than preoperatively. In the von Frey test the postoperative measurements were lower in the ipsilateral hind paw than preoperatively, while the contralateral hind paw showed an increase in maximal force before withdrawal. The contralateral hind paw showed more difference with sham mice than the ipsilateral hind paw. Silk ligatures showed more stable neuropathic pain behavior. In the acetone test, the cold plate test and the von Frey test the mice scored higher on neuropathic pain having silk ligatures, compared with catgut ligatures. Conclusion In this study we validated a murine CCI model for neuropathic pain behavior. In the murine CCI model it appears that silk ligatures demonstrate more stable neuropathic pain behaviors than catgut ligatures in de CCI model. Electronic supplementary material The online version of this article (doi:10.1186/s40064-015-1009-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Selina van der Wal
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Nijmegen Medical Centre (RUNMC), 6525 GA, Nijmegen, the Netherlands
| | - Lisa Cornelissen
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Nijmegen Medical Centre (RUNMC), 6525 GA, Nijmegen, the Netherlands
| | - Marije Behet
- Department of Microbiology RUNMC, 6525 GA, Nijmegen, the Netherlands
| | - Michiel Vaneker
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Nijmegen Medical Centre (RUNMC), 6525 GA, Nijmegen, the Netherlands
| | - Monique Steegers
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Nijmegen Medical Centre (RUNMC), 6525 GA, Nijmegen, the Netherlands
| | - Kris Vissers
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Nijmegen Medical Centre (RUNMC), 6525 GA, Nijmegen, the Netherlands
| |
Collapse
|
337
|
Downs CA, Faulkner MS. Toxic stress, inflammation and symptomatology of chronic complications in diabetes. World J Diabetes 2015; 6:554-565. [PMID: 25987953 PMCID: PMC4434076 DOI: 10.4239/wjd.v6.i4.554] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/30/2014] [Accepted: 02/12/2015] [Indexed: 02/05/2023] Open
Abstract
Diabetes affects at least 382 million people worldwide and the incidence is expected to reach 592 million by 2035. The incidence of diabetes in youth is skyrocketing as evidenced by a 21% increase in type 1 diabetes and a 30.5% increase in type 2 diabetes in the United States between 2001 and 2009. The effects of toxic stress, the culmination of biological and environmental interactions, on the development of diabetes complications is gaining attention. Stress impacts the hypothalamus-pituitary-adrenal axis and contributes to inflammation, a key biological contributor to the pathogenesis of diabetes and its associated complications. This review provides an overview of common diabetic complications such as neuropathy, cognitive decline, depression, nephropathy and cardiovascular disease. The review also provides a discussion of the role of inflammation and stress in the development and progression of chronic complications of diabetes, associated symptomatology and importance of early identification of symptoms of depression, fatigue, exercise intolerance and pain.
Collapse
|
338
|
Guedon JMG, Wu S, Zheng X, Churchill CC, Glorioso JC, Liu CH, Liu S, Vulchanova L, Bekker A, Tao YX, Kinchington PR, Goins WF, Fairbanks CA, Hao S. Current gene therapy using viral vectors for chronic pain. Mol Pain 2015; 11:27. [PMID: 25962909 PMCID: PMC4446851 DOI: 10.1186/s12990-015-0018-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/01/2015] [Indexed: 02/07/2023] Open
Abstract
The complexity of chronic pain and the challenges of pharmacotherapy highlight the importance of development of new approaches to pain management. Gene therapy approaches may be complementary to pharmacotherapy for several advantages. Gene therapy strategies may target specific chronic pain mechanisms in a tissue-specific manner. The present collection of articles features distinct gene therapy approaches targeting specific mechanisms identified as important in the specific pain conditions. Dr. Fairbanks group describes commonly used gene therapeutics (herpes simplex viral vector (HSV) and adeno-associated viral vector (AAV)), and addresses biodistribution and potential neurotoxicity in pre-clinical models of vector delivery. Dr. Tao group addresses that downregulation of a voltage-gated potassium channel (Kv1.2) contributes to the maintenance of neuropathic pain. Alleviation of chronic pain through restoring Kv1.2 expression in sensory neurons is presented in this review. Drs Goins and Kinchington group describes a strategy to use the replication defective HSV vector to deliver two different gene products (enkephalin and TNF soluble receptor) for the treatment of post-herpetic neuralgia. Dr. Hao group addresses the observation that the pro-inflammatory cytokines are an important shared mechanism underlying both neuropathic pain and the development of opioid analgesic tolerance and withdrawal. The use of gene therapy strategies to enhance expression of the anti-pro-inflammatory cytokines is summarized. Development of multiple gene therapy strategies may have the benefit of targeting specific pathologies associated with distinct chronic pain conditions (by Guest Editors, Drs. C. Fairbanks and S. Hao).
Collapse
Affiliation(s)
- Jean-Marc G Guedon
- Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA. .,Department of Ophthalmology, University of Pittsburgh School of Medicine, Room 1020 EEI, 203 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA.
| | - Xuexing Zheng
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | | | - Joseph C Glorioso
- Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 424 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Ching-Hang Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Shue Liu
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA.
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, State University of New Jersey, 185 S. Orange Ave., MSB, F-548, Newark, NJ, 07103, USA. .,Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA. .,Department of Neurology & Neuroscience, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA. .,Department of Physiology & Pharmacology, New Jersey Medical School, Rutgers, State University of New Jersey, Newark, NJ, 07103, USA.
| | - Paul R Kinchington
- Graduate Program in Molecular Virology and Microbiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA. .,Department of Ophthalmology, University of Pittsburgh School of Medicine, Room 1020 EEI, 203 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - William F Goins
- Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 424 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA, 15219, USA.
| | - Carolyn A Fairbanks
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA. .,Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA. .,Department of Pharmacology, University of Minnesota, 9-177 Weaver Densford Hall, 308 Harvard Street, Minneapolis, MN, 55455, USA.
| | - Shuanglin Hao
- Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
339
|
Zhang Q, Yu J, Wang J, Ding CP, Han SP, Zeng XY, Wang JY. The Red Nucleus TNF-α Participates in the Initiation and Maintenance of Neuropathic Pain Through Different Signaling Pathways. Neurochem Res 2015; 40:1360-71. [DOI: 10.1007/s11064-015-1599-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/11/2015] [Accepted: 05/02/2015] [Indexed: 11/30/2022]
|
340
|
Sun JS, Yang YJ, Zhang YZ, Huang W, Li ZS, Zhang Y. Minocycline attenuates pain by inhibiting spinal microglia activation in diabetic rats. Mol Med Rep 2015; 12:2677-82. [PMID: 25955348 DOI: 10.3892/mmr.2015.3735] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/26/2015] [Indexed: 11/05/2022] Open
Abstract
The mechanisms associated with diabetes-induced neuropathic pain are complex and poorly understood. In order to understand the involvement of spinal microglia activity in diabetic pain, the present study investigated whether minocycline treatment is able to attenuate diabetic pain using a rat model. Diabetes was induced using a single intraperitoneal injection of streptozotocin (STZ). Minocycline was then intrathecally administered to the rats. Paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were tested weekly. The expression of OX-42, Iba-1, phospho-p38 mitogen-activated protein kinase (MAPK), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and inducible nitric oxide synthase (iNOS), were examined in the spinal cord in order to evaluate the activation of microglia. The present study demonstrated that rats with STZ-induced diabetes exhibited increased mean plasma glucose concentration, decreased mean body weight and significant pain hypersensitivity compared with control rats. PWT and PWL values of rats with STZ-induced diabetes increased following treatment with minocycline. No differences were observed in expression levels of the microglial activity markers (OX-42, Iba-1 and phospho-p38 MAPK) between rats with STZ-induced diabetes and control rats. However, TNF-α, IL-1β and iNOS expression levels were higher in rats with STZ-induced diabetes compared with control rats. Following treatment with minocycline markers of microglial activation, including cytokines and iNOS, were downregulated in rats with STZ-induced diabetes. The results of the present study indicated that minocycline treatment may inhibit spinal microglial activation and attenuate diabetic pain in rats with STZ-induced diabetes.
Collapse
Affiliation(s)
- Jin-Shan Sun
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yu-Jie Yang
- Department of Ophthalmology, No. 474 Hospital of Chinese People's Liberation Army, Urumqi, Xinjiang 830013, P.R. China
| | - Yong-Zhen Zhang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Wen Huang
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, P.R. China
| | - Yong Zhang
- Department of Anatomy and K. K. Leung Brain Research Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
341
|
Miao GS, Liu ZH, Wei SX, Luo JG, Fu ZJ, Sun T. Lipoxin A4 attenuates radicular pain possibly by inhibiting spinal ERK, JNK and NF-κB/p65 and cytokine signals, but not p38, in a rat model of non-compressive lumbar disc herniation. Neuroscience 2015; 300:10-8. [PMID: 25943485 DOI: 10.1016/j.neuroscience.2015.04.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/08/2015] [Accepted: 04/27/2015] [Indexed: 01/12/2023]
Abstract
Inflammatory response induced by protrused nucleus pulposus (NP) has been shown to play a crucial role in the process of radicular pain. Lipoxins represent a unique class of lipid mediators that have anti-inflammatory and pro-resolving action. The present study was undertaken to investigate if intrathecal lipoxin A4 (LXA4) could alleviate mechanical allodynia in the rat models of application of NP to the L5 dorsal root ganglion (DRG). Non-compressive models of application of NP to L5 DRG were established and intrathecal catheterization for drug administration was performed in rats. Daily intrathecal injection of vehicle or LXA4 (10ng or 100ng) was performed for three successive days post-operation. Mechanical thresholds were tested and the ipsilateral lumbar (L4-L6) segment of spinal dorsal horns were removed for the determination of tumor necrosis factor-α (TNF-α), IL-1β, transforming growth factor-β1 (TGF-β1) and IL-10 expression and NF-κB/p65, extracellular signal-regulated kinase (ERK), C-Jun N-terminal kinase (JNK) and P38 expression. Application of NP to DRG in rats induced mechanical allodynia, increased the expression of pro-inflammatory factors (TNF-α and IL-1β), NF-κB/p65, the phosphorylated-ERK (p-ERK), -JNK (p-JNK) and -P38 (p-p38) and decreased the expression of anti-inflammatory cytokines (TGF-β1 and IL-10) in the ipsilateral lumbar (L4-L6) segment of spinal dorsal horns. Intrathecal injection of LXA4 alleviated the development of neuropathic pain, inhibited the upregulation of pro-inflammatory cytokines (TNF-α and IL-1β), upregulated the expression of anti-inflammatory cytokines (TGF-β1 and IL-10) and attenuated the activation of NF-κB/p65, p-ERK, p-JNK, but not p-p38, in a dose-dependent manner. In this study, we have demonstrated that LXA4 potently alleviate radicular pain in a rat model of non-compressive lumbar disc herniation. The anti-inflammatory and pro-resolution properties of LXA4 have shown a great promise for the management of radicular pain caused by intervertebral disc herniation.
Collapse
Affiliation(s)
- G-S Miao
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China
| | - Z-H Liu
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China
| | - S-X Wei
- Department of Anesthesiology, The Sixth People's Hospital of Jinan, Zhangqiu, Shandong, PR China
| | - J-G Luo
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China
| | - Z-J Fu
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China
| | - T Sun
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, PR China.
| |
Collapse
|
342
|
Shipton EE, Shipton EA. Vitamin D Deficiency and Pain: Clinical Evidence of Low Levels of Vitamin D and Supplementation in Chronic Pain States. Pain Ther 2015; 4:67-87. [PMID: 25920326 PMCID: PMC4470966 DOI: 10.1007/s40122-015-0036-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Indexed: 12/31/2022] Open
Abstract
Introduction A number of studies suggest a link between low levels of 25-hydroxy vitamin D and incidence of acute and chronic pain. Clinical studies of vitamin D supplementation in patients with known vitamin D deficiency have shown mixed results in improving pain scores. Methods In this article, vitamin D deficiency risk factors are observed and adequate levels of 25-hydroxy vitamin D defined. Clinical supplementation with vitamin D is explored, including the schedules used in published clinical trials. Evidence of the effectiveness of vitamin D supplementation for the treatment of chronic pain conditions from double-blind randomized controlled trials (RCTs) is examined. Results The scientific evidence for vitamin D as a treatment option for chronic pain is limited due to lack of RCTs. It cannot be stated conclusively that vitamin D deficiency is directly linked to the etiology or maintenance of chronic pain states. Conclusion There remains a growing body of both clinical and laboratory evidence pointing to a potential relationship between low levels of 25-hydroxy vitamin D and a variety of chronic pain states. More focused research involving large RCTs is necessary. Electronic supplementary material The online version of this article (doi:10.1007/s40122-015-0036-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elspeth E Shipton
- Department of Anaesthesia, University of Otago, Christchurch, New Zealand
| | | |
Collapse
|
343
|
Vitamin D and Pain: Vitamin D and Its Role in the Aetiology and Maintenance of Chronic Pain States and Associated Comorbidities. PAIN RESEARCH AND TREATMENT 2015; 2015:904967. [PMID: 26090221 PMCID: PMC4427945 DOI: 10.1155/2015/904967] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/15/2015] [Indexed: 12/25/2022]
Abstract
The emergence of new data suggests that the benefits of Vitamin D extend beyond healthy bones. This paper looks at Vitamin D and its role in the aetiology and maintenance of chronic pain states and associated comorbidities. The interfaces between pain and Vitamin D and the mechanisms of action of Vitamin D on pain processes are explored. Finally the association between Vitamin D and pain comorbidities such as sleep and depression is investigated. The paper shows that Vitamin D exerts anatomic, hormonal, neurological, and immunological influences on pain manifestation, thereby playing a role in the aetiology and maintenance of chronic pain states and associated comorbidities. More research is necessary to determine whether Vitamin D is useful in the treatment of various pain conditions and whether or not the effect is limited to patients who are deficient in Vitamin D.
Collapse
|
344
|
Sun Y, Yang M, Tang H, Ma Z, Liang Y, Li Z. The over-production of TNF-α via Toll-like receptor 4 in spinal dorsal horn contributes to the chronic postsurgical pain in rat. J Anesth 2015; 29:734-40. [PMID: 25895164 DOI: 10.1007/s00540-015-2011-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 03/29/2015] [Indexed: 11/29/2022]
Abstract
PURPOSE Many patients suffer from chronic postsurgical pain (CPSP) following surgery, and the underlying mechanisms are poorly understood. In the present work, using the skin/muscle incision retraction (SMIR) model, the role of spinal TLR4/TNF-α pathway in the induction of CPSP was evaluated. METHODS Mechanical allodynia induced by SMIR was established in adult male Sprague-Dawley rats. The von Frey test was performed to evaluate the role of TLR4/TNF-α pathway on the mechanical allodynia. Western-blot and immunohistochemistry methods were adopted to understand the molecular mechanisms. RESULTS SMIR surgery decreased the ipsilateral 50 % paw withdrawal threshold, lasting for at least 20 days. Western-blot analysis and immunohistochemistry revealed that SMIR surgery significantly upregulated the expression of TLR4 (p < 0.01) in glial cells on the ipsilateral side of spinal cord and increased TLR4 occurred on day 5 and was maintained to the end of the experiment (day 20). Similarly, tumor necrosis factor-alpha (TNF-α) was significantly increased on days 5, 10, and 20 on the ipsilateral side of spinal dorsal horn following SMIR surgery. Intraperitoneal injection of an inhibitor of TNF-α synthesis thalidomide at 50 or 100 mg/kg dose (but not 10 mg/kg dose) significantly ameliorated the reduced paw withdrawal threshold induced by SMIR surgery. Importantly, intrathecal delivery of a specific TLR4 antagonist (LPS-RS) at dose of 25 μg significantly attenuated mechanical allodynia and prevented the upregulation of TNF-α induced by SMIR surgery. CONCLUSIONS These findings suggest that the upregulation of TNF-α via TLR4 contributes to the development of CPSP in spinal dorsal horn.
Collapse
Affiliation(s)
- Yang Sun
- Department of Pain, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Xinjiang, 830054, China
| | - Mingmin Yang
- Department of Pain, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, Xinjiang, 830054, China
| | - Hao Tang
- Department of General Internal Medicine, The First Affiliated Hospital of Sun Yat-sen University, Sun Yet-Sen University, 58 Zhongshan Rd. 2, Guangzhou, 510080, China
| | - Zhongfu Ma
- Department of General Internal Medicine, The First Affiliated Hospital of Sun Yat-sen University, Sun Yet-Sen University, 58 Zhongshan Rd. 2, Guangzhou, 510080, China
| | - Yanbing Liang
- Department of General Internal Medicine, The First Affiliated Hospital of Sun Yat-sen University, Sun Yet-Sen University, 58 Zhongshan Rd. 2, Guangzhou, 510080, China
| | - Zhenyu Li
- Department of General Internal Medicine, The First Affiliated Hospital of Sun Yat-sen University, Sun Yet-Sen University, 58 Zhongshan Rd. 2, Guangzhou, 510080, China.
| |
Collapse
|
345
|
Hawkins JL, Denson JE, Miley DR, Durham PL. Nicotine stimulates expression of proteins implicated in peripheral and central sensitization. Neuroscience 2015; 290:115-25. [PMID: 25637801 PMCID: PMC5894823 DOI: 10.1016/j.neuroscience.2015.01.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/19/2014] [Accepted: 01/07/2015] [Indexed: 12/26/2022]
Abstract
Pain patients who are nicotine dependent report a significantly increased incidence and severity of pain intensity. The goal of this study was to determine the effects of prolonged nicotine administration on inflammatory proteins implicated in the development of peripheral and central sensitization of the trigeminal system. Behavioral, immunohistochemical, and microarray studies were utilized to investigate the effects of nicotine administered daily for 14 days via an Alzet® osmotic pump in Sprague Dawley rats. Systemic nicotine administration caused a significant increase in nocifensive withdrawals to mechanical stimulation of trigeminal neurons. Nicotine stimulated expression of the pro-inflammatory signal transduction proteins phosphorylated-extracellular signal-regulated kinase (p-ERK), phosphorylated-c-Jun N-terminal kinase (p-JNK), and protein kinase A (PKA) in the spinal trigeminal nucleus. Nicotine also promoted elevations in the expression of glial fibrillary acidic protein (GFAP), a biomarker of activated astrocytes, and the microglia biomarker ionized calcium-binding adapter molecule 1 (Iba1). Similarly, levels of eleven cytokines were significantly elevated with the largest increase in expression of TNF-α. Levels of PKA, p-ERK, and p-JNK in trigeminal ganglion neurons were increased by nicotine. Our findings demonstrate that prolonged systemic administration of nicotine promotes sustained behavioral and cellular changes in the expression of key proteins in the spinal trigeminal nucleus and trigeminal ganglion implicated in the development and maintenance of peripheral and central sensitization.
Collapse
Affiliation(s)
- J L Hawkins
- Center for Biomedical & Life Sciences, Missouri State University, Springfield, MO, USA
| | - J E Denson
- Center for Biomedical & Life Sciences, Missouri State University, Springfield, MO, USA
| | - D R Miley
- Center for Biomedical & Life Sciences, Missouri State University, Springfield, MO, USA
| | - P L Durham
- Center for Biomedical & Life Sciences, Missouri State University, Springfield, MO, USA.
| |
Collapse
|
346
|
Transient Hyperesthesia and Radicular Pain: An Unrecognised Complication of Thoracic Radiotherapy. J Thorac Oncol 2015; 10:708-12. [DOI: 10.1097/jto.0000000000000469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
347
|
Galluzzo M, Talamonti M, Di Stefani A, Chimenti S. Linear psoriasis following the typical distribution of the sciatic nerve. J Dermatol Case Rep 2015; 9:6-11. [PMID: 25932056 DOI: 10.3315/jdcr.2015.1189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 01/04/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND Some studies suggest that the nervous system plays a role in the onset of psoriasis and psoriasis flares including the symmetry of lesions, sparing of denervated skin and the role of stress in inducing lesions. MAIN OBSERVATIONS We describe an unusual case of psoriasis occurring in the same distribution as sciatic pain from a prolapsed intervertebral disc. The patient, a 45-year-old man with plaque psoriasis was treated with ustekinumab for 104 weeks, at a standard dose. During the eight month of therapy he developed an asymptomatic linear eruption on the left lower extremity along the distribution of the sciatic nerve. On examination, erythematous scaly plaques were noted. Histopathology confirmed the diagnosis of psoriasis. The treatment was continued and clobetasol proprionate 0.05% cream was added. At week 12 after the eruption, the patient reported a pain radiating through the buttock and posterior left leg during jogging. Magnetic resonance imaging showed lumbar disc herniation with compression of the L5-S1 spinal nerve roots. The patient stopped running and the psoriasis spontaneously receded, in a slow but complete fashion, without any local treatment. CONCLUSION There is substantial evidence that nerves play a key role in the pathogenesis of psoriasis. We hypothesized that local TNF-alpha, neuropeptides and nerve growth factor, which are produced by nerve root compression, played a critical role in this case of psoriasis onset in an area of pain from a bulging lumbar intervertebral disc. To our knowledge, a correlation of psoriasis and nerve root compression has not been described previously.
Collapse
Affiliation(s)
- Marco Galluzzo
- Department of Dermatology, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy
| | - Marina Talamonti
- Department of Dermatology, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy
| | - Alessandro Di Stefani
- Department of Dermatology, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy
| | - Sergio Chimenti
- Department of Dermatology, University of Rome "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy
| |
Collapse
|
348
|
Choi BM, Lee SH, An SM, Park DY, Lee GW, Noh GJ. The time-course and RNA interference of TNF-α, IL-6, and IL-1β expression on neuropathic pain induced by L5 spinal nerve transection in rats. Korean J Anesthesiol 2015; 68:159-69. [PMID: 25844135 PMCID: PMC4384404 DOI: 10.4097/kjae.2015.68.2.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 11/23/2014] [Accepted: 12/12/2014] [Indexed: 01/09/2023] Open
Abstract
Background The objective of this study was to investigate the time-course of the expression of TNF-α, IL-6, and IL-1β after L5 spinal nerve transection (SNT), and to determine the effect of small interfering RNA (siRNA) targeting these cytokines on neuropathic pain. Methods Rats received control siRNA (CON group, n = 80) or a cocktail of siRNAs targeting these cytokines (COCK group, n = 70). The siRNAs were given via intrathecal catheter 1 d prior to SNT, on the operation day, and 1, 2 and 3 d postoperatively. Behavioral tests and levels of the cytokine mRNAs and proteins as well as glial cell activity were following the L5 SNT. Results In the CON group, TNF-α and IL-1β mRNA levels increased immediately after SNT and remained high for 6 d, while IL-6 transcripts only began to increase after 12 h. TNF-α and IL-1β mRNA levels in the COCK group were lower than in the CON group at all time points (P < 0.05). In the behavioral tests, allodynia and hyperalgesia were significantly lower in the COCK group from 2 d after SNT (P < 0.05). Conclusions The time courses of TNF-α, IL-6 and IL-1β mRNA expression after L5 SNT differ. RNA interference may be a method of reducing the development of mechanical allodynia and hyperalgesia in response to nerve injury.
Collapse
Affiliation(s)
- Byung Moon Choi
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Han Lee
- Department of Clinical Pharmacology and Therapeutics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Mee An
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Do Yang Park
- Department of Clinical Pharmacology and Therapeutics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gwan Woo Lee
- Department of Anesthesiology and Pain Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Gyu-Jeong Noh
- Department of Anesthesiology and Pain Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. ; Department of Clinical Pharmacology and Therapeutics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
349
|
Huang SY, Sung CS, Chen WF, Chen CH, Feng CW, Yang SN, Hung HC, Chen NF, Lin PR, Chen SC, Wang HMD, Chu TH, Tai MH, Wen ZH. Involvement of phosphatase and tensin homolog deleted from chromosome 10 in rodent model of neuropathic pain. J Neuroinflammation 2015; 12:59. [PMID: 25889774 PMCID: PMC4386079 DOI: 10.1186/s12974-015-0280-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/07/2015] [Indexed: 12/30/2022] Open
Abstract
Background Many cancer research studies have extensively examined the phosphatase and tensin homolog deleted from chromosome 10 (PTEN) pathway. There are only few reports that suggest that PTEN might affect pain; however, there is still a lack of evidence to show the role of PTEN for modulating pain. Here, we report a role for PTEN in a rodent model of neuropathic pain. Results We found that chronic constriction injury (CCI) surgery in rats could elicit downregulation of spinal PTEN as well as upregulation of phosphorylated PTEN (phospho-PTEN) and phosphorylated mammalian target of rapamycin (phospho-mTOR). After examining such changes in endogenous PTEN in neuropathic rats, we explored the effects of modulating the spinal PTEN pathway on nociceptive behaviors. The normal rats exhibited mechanical allodynia after intrathecal (i.t.) injection of adenovirus-mediated PTEN antisense oligonucleotide (Ad-antisense PTEN). These data indicate the importance of downregulation of spinal PTEN for nociception. Moreover, upregulation of spinal PTEN by i.t. adenovirus-mediated PTEN (Ad-PTEN) significantly prevented CCI-induced development of nociceptive sensitization, thermal hyperalgesia, mechanical allodynia, cold allodynia, and weight-bearing deficits in neuropathic rats. Furthermore, upregulation of spinal PTEN by i.t. Ad-PTEN significantly attenuated CCI-induced microglia and astrocyte activation, upregulation of tumor necrosis factor-α (TNF-α) and phospho-mTOR, and downregulation of PTEN in neuropathic rats 14 days post injury. Conclusions These findings demonstrate that PTEN plays a key, beneficial role in a rodent model of neuropathic pain.
Collapse
Affiliation(s)
- Shi-Ying Huang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Center for Neuroscience, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Road, Taipei, 11217, Taiwan. .,School of Medicine, National Yang-Ming University, No. 155, Section 2, Linong Street, Taipei, 11221, Taiwan.
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No. 123, DAPI Road, Kaohsiung, 83301, Taiwan. .,Department of Neurosurgery, Xiamen Chang Gung Memorial Hospital, No. 123, Xiafei Road, Fujian, 361026, China.
| | - Chun-Hong Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Chien-Wei Feng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - San-Nan Yang
- School of Medicine, College of Medicine and Department of Pediatrics, E-DA Hospital, I-Shou University, No. 1, Yida Road, Kaohsiung, 82445, Taiwan.
| | - Han-Chun Hung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Nan-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, No. 2, Zhongzheng 1st Road, Kaohsiung, 80284, Taiwan.
| | - Pey-Ru Lin
- Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - San-Cher Chen
- Center for Neuroscience, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Hui-Min David Wang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Kaohsiung, 80708, Taiwan. .,Graduate Institute of Natural Products, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Kaohsiung, 80708, Taiwan. .,Center for Stem Cell Research, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Kaohsiung, 80708, Taiwan.
| | - Tian-Huei Chu
- Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Ming-Hong Tai
- Center for Neuroscience, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Institute of Biomedical Sciences, National Sun Yat-sen University, #70 Lienhai Road, Kaohsiung, 80424, Taiwan. .,Department of Biological Sciences, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan. .,Marine Biomedical Laboratory and Center for Translational Biopharmaceuticals, Department of Marine Biotechnology and Resources, National Sun Yat-sen University, No. 70, Lienhai Road, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
350
|
Salvat E, Schweitzer B, Massard G, Meyer N, de Blay F, Muller A, Barrot M. Effects of β2 agonists on post-thoracotomy pain incidence. Eur J Pain 2015; 19:1428-36. [PMID: 25766791 DOI: 10.1002/ejp.673] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Pre-clinical research has shown β2 -adrenoceptors to be essential for the antiallodynic action of antidepressant drugs in murine models of neuropathic pain and that sustained treatment with β2 -agonists has an antiallodynic action. Here, we clinically investigated whether chronic β2 -agonist treatments may influence the incidence of post-thoracotomy chronic pain, defined as pain that recurs or persists along a thoracotomy scar more than 2 months after surgery, either neuropathic or non-neuropathic. METHODS We conducted an epidemiological study on patients operated by thoracotomy. Demographic data, medical history and treatments concomitant to the surgery were recorded at a follow-up visit. Information on perioperative treatments was collected from the anaesthesia records and confirmed by the patients. In patients with pain at the surgery level, post-thoracotomy chronic pain was assessed by clinical examination and numeric scale. Physical examination and DN4 questionnaire were used to discriminate neuropathic and non-neuropathic chronic pain at scar level. RESULTS One hundred and eighty-nine patients were included. Eighty-one patients reported persisting thoracic pain, with neuropathic characteristics in 58 of them (30% of the 189 patients). The most common chronic drugs during the perioperative period were inhaled β2 -agonists (28.6%). The chronic use of β2 -agonists was an independent predictor of thoracic neuropathic pain (but not of non-neuropathic pain) and was associated with a five-fold decrease in the relative incidence of neuropathic pain [OR = 0.19 (0.06-0.45)]. CONCLUSIONS These data suggest a possible influence of chronic β2 -agonist treatments on neuropathic pain secondary to thoracotomy. This apparent preventive effect of β2 -agonist treatments should warrant controlled clinical trials.
Collapse
Affiliation(s)
- E Salvat
- Centre d'Evaluation et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - B Schweitzer
- Département d'Anesthésie-Réanimation Chirurgicale, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - G Massard
- Département de Pathologies Thoraciques, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - N Meyer
- Service de Santé Publique, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - F de Blay
- Département de Pathologies Thoraciques, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - A Muller
- Centre d'Evaluation et de Traitement de la Douleur, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - M Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, France
| |
Collapse
|