1
|
Ho MFS, Farkas O, Faria AV, Plemel JR, Kerr BJ. A recent history of immune cell sex differences in the peripheral nervous system in persistent pain states. Brain Behav Immun 2025:S0889-1591(25)00181-3. [PMID: 40345628 DOI: 10.1016/j.bbi.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025] Open
Abstract
Pain is entwined with inflammation, and biological sex often influences mechanisms of the immune system. Due to possible differences in inflammatory mechanisms, women are predisposed to autoimmune diseases and chronic pain. Despite sex as a critical variable in clinical cases of autoimmune conditions and its pain comorbidities, fundamental investigations have long underrepresented female subjects in their studies. Fundamental research in the 2010 s, however, identified a binary sex specific mechanism for pain in rodents: male pain is microglia-driven while female pain is T cell-driven. Since then, studies have expanded in neuro-immunology to indicate that the sex differences and immune cells involved in these processes take on more elaborate roles when expanded to other causal modalities and anatomical levels of neuropathic and inflammatory pain. In this mini-review, we highlight updated roles for macrophages, T cells, and B cells in the peripheral nervous system during persistent pain conditions: neuropathic pain and inflammatory pain. We discuss sex similarities and sex differences in these cell types. By parsing out the sex specific roles of immune cells in persistent pain states we may be better positioned to find immune-based therapies that can effectively target chronic pain in sex-biased autoimmune conditions.
Collapse
Affiliation(s)
| | - Olivia Farkas
- Neuroscience and Mental Health Institute, University of Alberta, Canada
| | | | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Canada; Department of Medicine, Division of Neurology, University of Alberta, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Canada; Departments of Pharmacology and Physiology, University of Alberta, Canada; Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
2
|
Tong SH, Liu DL, Liao P, Zhang SY, Zhou J, Zong Y, Zhang CQ, Huang YG, Gao JJ. Emerging role of macrophages in neuropathic pain. J Orthop Translat 2025; 51:227-241. [PMID: 40177638 PMCID: PMC11964759 DOI: 10.1016/j.jot.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 04/05/2025] Open
Abstract
Neuropathic pain is a complex syndrome caused by injury to the neurons, which causes persistent hypersensitivity and considerable inconvenience to the patient's whole life. Over the past two decades, the interaction between immune cells and neurons has been proven to play a crucial role in the development of neuropathic pain. Increasing studies have indicated the important role of macrophages for neuroinflammation and have shed light on the underlying molecular and cellular mechanisms. In addition, novel therapeutic methods targeting macrophages are springing up, which provide more options in our clinical treatment. Herein, we reviewed the characteristics of peripheral macrophages and their function in neuropathic pain, with the aim of better understanding how these cells contribute to pathological processes and paving the way for therapeutic approaches. Translational potential statement This review provides a comprehensive overview of the mechanisms underlying the interplay between the macrophages and nervous system during the progression of nerve injury. Additionally, it compiles existing intervention strategies targeting macrophages for the treatment of neuropathic pain. This information offers valuable insights for researchers seeking to address the challenge of this intractable pain.
Collapse
Affiliation(s)
- Si-Han Tong
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - De-Lin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Sen-Yao Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jian Zhou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, 6009, Australia
| | - Chang-Qing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yi-Gang Huang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jun-Jie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
3
|
Zamith Cunha R, Semprini A, Salamanca G, Gobbo F, Morini M, Pickles KJ, Roberts V, Chiocchetti R. Expression of Cannabinoid Receptors in the Trigeminal Ganglion of the Horse. Int J Mol Sci 2023; 24:15949. [PMID: 37958932 PMCID: PMC10648827 DOI: 10.3390/ijms242115949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Cannabinoid receptors are expressed in human and animal trigeminal sensory neurons; however, the expression in the equine trigeminal ganglion is unknown. Ten trigeminal ganglia from five horses were collected post-mortem from an abattoir. The expression of cannabinoid receptors type 1 (CB1R) and type 2 (CB2R), and the cannabinoid-related receptors like transient receptor potential vanilloid type 1 (TRPV1), peroxisome proliferator-activated receptor gamma (PPARɣ), and G protein-related receptor 55 (GPR55) in the trigeminal ganglia (TG) of the horse were studied, using immunofluorescence on cryosections and formalin-fixed paraffin-embedded (FFPE) sections. Neurons and glial cells were identified using fluorescent Nissl staining NeuroTrace® and an antibody directed against the glial marker glial fibrillary acidic protein (GFAP), respectively. Macrophages were identified by means of an antibody directed against the macrophages/microglia marker ionized calcium-binding adapter molecule 1 (IBA1). The protein expression of CB1R, CB2R, TRPV1, and PPARɣ was found in the majority of TG neurons in both cryosections and FFPE sections. The expression of GPR55 immunoreactivity was mainly detectable in FFPE sections, with expression in the majority of sensory neurons. Some receptors were also observed in glial cells (CB2R, TRPV1, PPARγ, and GPR55) and inflammatory cells (PPARγ and GPR55). These results support further investigation of such receptors in disorders of equine trigeminal neuronal excitability.
Collapse
Affiliation(s)
- Rodrigo Zamith Cunha
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Alberto Semprini
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Giulia Salamanca
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Maria Morini
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Kirstie J. Pickles
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham LE12 5RD, UK;
| | - Veronica Roberts
- Bristol Vet School, University of Bristol, Bristol BS40 5DU, UK;
| | - Roberto Chiocchetti
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| |
Collapse
|
4
|
Abstract
Interactions between the immune and nervous systems are of central importance in neuropathic pain, a common and debilitating form of chronic pain caused by a lesion or disease affecting the somatosensory system. Our understanding of neuroimmune interactions in pain research has advanced considerably. Initially considered as passive bystanders, then as culprits in the pathogenesis of neuropathic pain, immune responses in the nervous system are now established to underpin not only the initiation and progression of pain but also its resolution. Indeed, immune cells and their mediators are well-established promoters of neuroinflammation at each level of the neural pain pathway that contributes to pain hypersensitivity. However, emerging evidence indicates that specific subtypes of immune cells (including antinociceptive macrophages, pain-resolving microglia and T regulatory cells) as well as immunoresolvent molecules and modulators of the gut microbiota-immune system axis can reduce the pain experience and contribute to the resolution of neuropathic pain. This Review provides an overview of the immune mechanisms responsible for the resolution of neuropathic pain, including those involved in innate, adaptive and meningeal immunity as well as interactions with the gut microbiome. Specialized pro-resolving mediators and therapeutic approaches that target these neuroimmune mechanisms are also discussed.
Collapse
|
5
|
Santos DFS, Donahue RR, Laird D, Oliveira M, Taylor B. The PPARγ agonist pioglitazone produces a female-predominant inhibition of hyperalgesia associated with surgical incision, peripheral nerve injury, and painful diabetic neuropathy. Neuropharmacology 2022; 205:108907. [PMID: 34856203 PMCID: PMC8992004 DOI: 10.1016/j.neuropharm.2021.108907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/12/2021] [Accepted: 11/28/2021] [Indexed: 12/21/2022]
Abstract
Pioglitazone, an agonist at peroxisome proliferator-activated receptor gamma, is FDA-approved for the treatment of insulin resistance in type 2 diabetes. Numerous studies in male rodents suggest that pioglitazone inhibits inflammatory and neuropathic pain, but few included female subjects. To address this gap, we compared the effects of pioglitazone in both sexes in the intraplantar methylglyoxal model (MG) model of chemical pain and painful diabetic neuropathy (PDN), the plantar incision model (PIM) of postoperative pain, the spared nerve injury (SNI) model of traumatic nerve injury, and the ZDF rat and db/db mouse models of PDN. We administered pioglitazone by one-time intrathecal or intraperitoneal injection or by adding it to chow for 6 weeks, followed by measurement of hypersensitivity to non-noxious mechanical, noxious mechanical, heat, and/or cold stimuli. In all mouse models, injection of pioglitazone decreased pain-like behaviors with greater potency and/or efficacy in females as compared to males: heat and mechanical hypersensitivity in the MG model (0.1-10 mg/kg); mechanical hypersensitivity in the PIM model (10 μg); mechanical and cold hypersensitivity in the SNI model (100 mg/kg); and heat hypersensitivity in the db/db model (100 mg/kg). Furthermore, co-administration of low doses of morphine (1 mg/kg) and pioglitazone (10 mg/kg) decreased SNI-induced mechanical and cold hypersensitivity in female but not male mice. In the ZDF rat, pioglitazone (100 mg/kg) decreased heat and mechanical hypersensitivity with no sex difference. In the db/db model, pioglitazone had no effect when given into chow for 6 weeks at 0.3, 3 or 30 mg/kg doses. We conclude that females exhibit greater anti-hyperalgesic responses to pioglitazone in mouse models of chemical-induced nociception, postsurgical pain, neuropathic pain, and PDN. These findings set the stage for clinical trials to determine whether pioglitazone has analgesic properties across a broad spectrum of chronic pain conditions, particularly in women.
Collapse
Affiliation(s)
- D. F. S. Santos
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA, USA.,School of Applied Sciences, State University of Campinas-UNICAMP, Limeira, SP, Brazil
| | - R. R. Donahue
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY, USA
| | - D.E. Laird
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY, USA
| | - M.C. Oliveira
- School of Applied Sciences, State University of Campinas-UNICAMP, Limeira, SP, Brazil
| | - B.K. Taylor
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, and Pittsburgh Project to end Opioid Misuse, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Silva NR, Gomes FIF, Lopes AHP, Cortez IL, Dos Santos JC, Silva CEA, Mechoulam R, Gomes FV, Cunha TM, Guimarães FS. The Cannabidiol Analog PECS-101 Prevents Chemotherapy-Induced Neuropathic Pain via PPARγ Receptors. Neurotherapeutics 2022; 19:434-449. [PMID: 34904193 PMCID: PMC9130439 DOI: 10.1007/s13311-021-01164-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 01/03/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is the main dose-limiting adverse effect of chemotherapy drugs such as paclitaxel (PTX). PTX causes marked molecular and cellular damage, mainly in the peripheral nervous system, including sensory neurons in the dorsal root ganglia (DRG). Several studies have shown the therapeutic potential of cannabinoids, including cannabidiol (CBD), the major non-psychotomimetic compound found in the Cannabis plant, to treat peripheral neuropathies. Here, we investigated the efficacy of PECS-101 (former HUF-101), a CBD fluorinated analog, on PTX-induced neuropathic pain in mice. PECS-101, administered after the end of treatment with PTX, did not reverse mechanical allodynia. However, PECS-101 (1 mg/kg) administered along with PTX treatment caused a long-lasting relief of the mechanical and cold allodynia. These effects were blocked by a PPARγ, but not CB1 and CB2 receptor antagonists. Notably, the effects of PECS-101 on the relief of PTX-induced mechanical and cold allodynia were not found in macrophage-specific PPARγ-deficient mice. PECS-101 also decreased PTX-induced increase in Tnf, Il6, and Aif1 (Iba-1) gene expression in the DRGs and the loss of intra-epidermal nerve fibers. PECS-101 did not alter motor coordination, produce tolerance, or show abuse potential. In addition, PECS-101 did not interfere with the chemotherapeutic effects of PTX. Thus, PECS-101, a new fluorinated CBD analog, could represent a novel therapeutic alternative to prevent mechanical and cold allodynia induced by PTX potentially through the activation of PPARγ in macrophages.
Collapse
Affiliation(s)
- Nicole Rodrigues Silva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | | | | | - Isadora Lopes Cortez
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Conceição Elidianne Aníbal Silva
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Raphael Mechoulam
- Department of Medicinal Chemistry and Natural Products, Hebrew University Medical Faculty, Jerusalem, Israel
| | - Felipe Villela Gomes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Francisco Silveira Guimarães
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
- National Institute of Science and Translational Medicine, Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
7
|
Hassan FE, Sakr HI, Mohie PM, Suliman HS, Mohamed AS, Attia MH, Eid DM. Pioglitazone improves skeletal muscle functions in reserpine-induced fibromyalgia rat model. Ann Med 2021; 53:1032-1040. [PMID: 34233552 PMCID: PMC8274527 DOI: 10.1080/07853890.2021.1916069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/24/2021] [Accepted: 04/03/2021] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Fibromyalgia (FM) is characterized by musculoskeletal pain, fatigue, sleep and memory disturbance. There is no definitive cure yet for FM-related health problems. Peroxisome proliferator-activated receptor's (PPAR's) activation is associated with insulin sensitisation and improved glucose metabolism. PPAR-γ was reported to alleviate FM allodynia. Limited data are discussing its effect on motor disorders. OBJECTIVE To investigate the potential effect of PPAR-γ agonists (pioglitazone, as one member of thiazolidinediones (TZD)) on motor dysfunction in reserpine-induced FM in a rat model. MATERIALS AND METHODS Thirty-six male Wistar rats were divided into negative control (n = 9) and reserpine-induced FM (n = 27) groups. The latter was subdivided into three equal subgroups (n = 9), positive control (untreated FM model), pioglitazone-treated and GW9662-treated. We evaluated muscle functions and activity of chloramphenicol acetyltransferase, superoxide dismutase, malondialdehyde, and serum levels of interleukin-8 and monocyte chemoattractant protein-1. RESULTS Pioglitazone significantly relieved fatigue, improved muscle performance, reduced inflammatory cytokines and enhanced antioxidant's activity, while GW9662, a known PPAR-γ antagonist, aggravated the FM manifestations in the rat model. CONCLUSION PPAR-γ agonists show a promising role against FM-associated motor dysfunctions.
Collapse
Affiliation(s)
- Fatma E. Hassan
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Hader I. Sakr
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Egypt
- Department of Medical Physiology, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Passant M. Mohie
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Egypt
| | - Howayda Saeed Suliman
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Egypt
| | | | - Mohamed H. Attia
- Internal Medicine Department, Faculty of Medicine, Ain Shams University, Egypt
| | - Dalia M. Eid
- Department of Biochemistry, Faculty of Medicine, Ain Shams University, Egypt
| |
Collapse
|
8
|
Rosiglitazone Alleviates Mechanical Allodynia of Rats with Bone Cancer Pain through the Activation of PPAR- γ to Inhibit the NF- κB/NLRP3 Inflammatory Axis in Spinal Cord Neurons. PPAR Res 2021; 2021:6086265. [PMID: 34484316 PMCID: PMC8413064 DOI: 10.1155/2021/6086265] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/28/2021] [Indexed: 02/08/2023] Open
Abstract
Bone cancer pain (BCP) is a serious clinical problem that affects the quality of life of cancer patients. However, the current treatment methods for this condition are still unsatisfactory. This study investigated whether intrathecal injection of rosiglitazone modulates the noxious behaviors associated with BCP, and the possible mechanisms related to this effect were explored. We found that rosiglitazone treatment relieved bone cancer-induced mechanical hyperalgesia in a dose-dependent manner, promoted the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) in spinal cord neurons, and inhibited the activation of the nuclear factor-kappa B (NF-κB)/nod-like receptor protein 3 (NLRP3) inflammatory axis induced by BCP. However, concurrent administration of the PPAR-γ antagonist GW9662 reversed these effects. The results show that rosiglitazone inhibits the NF-κB/NLRP3 inflammation axis by activating PPAR-γ in spinal neurons, thereby alleviating BCP. Therefore, the PPAR-γ/NF-κB/NLRP3 signaling pathway may be a potential target for the treatment of BCP in the future.
Collapse
|
9
|
Bethea JR, Fischer R. Role of Peripheral Immune Cells for Development and Recovery of Chronic Pain. Front Immunol 2021; 12:641588. [PMID: 33692810 PMCID: PMC7937804 DOI: 10.3389/fimmu.2021.641588] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/03/2021] [Indexed: 01/01/2023] Open
Abstract
Chronic neuropathic pain (CNP) is caused by a lesion or disease of the somatosensory nervous system. It affects ~8% of the general population and negatively impacts a person's level of functioning and quality of life. Its resistance to available pain therapies makes CNP a major unmet medical need. Immune cells have been shown to play a role for development, maintenance and recovery of CNP and therefore are attractive targets for novel pain therapies. In particular, in neuropathic mice and humans, microglia are activated in the dorsal horn and peripheral immune cells infiltrate the nervous system to promote chronic neuroinflammation and contribute to the initiation and progression of CNP. Importantly, immunity not only controls pain development and maintenance, but is also essential for pain resolution. In particular, regulatory T cells, a subpopulation of T lymphocytes with immune regulatory function, and macrophages were shown to be important contributors to pain recovery. In this review we summarize the interactions of the peripheral immune system with the nervous system and outline their contribution to the development and recovery of pain.
Collapse
Affiliation(s)
- John R. Bethea
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Roman Fischer
- Institute of Cell Biology and Immunology, University Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
10
|
Machelska H, Celik MÖ. Immune cell-mediated opioid analgesia. Immunol Lett 2020; 227:48-59. [PMID: 32814155 DOI: 10.1016/j.imlet.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022]
Abstract
Pathological pain is regulated by a balance between pro-algesic and analgesic mechanisms. Interactions between opioid peptide-producing immune cells and peripheral sensory neurons expressing opioid receptors represent a powerful intrinsic pain control in animal models and in humans. Therefore, treatments based on general suppression of immune responses have been mostly unsuccessful. It is highly desirable to develop strategies that specifically promote neuro-immune communication mediated by opioids. Promising examples include vaccination-based recruitment of opioid-containing leukocytes to painful tissue and the local reprogramming of pro-algesic immune cells into analgesic cells producing and secreting high amounts of opioid peptides. Such approaches have the potential to inhibit pain at its origin and be devoid of central and systemic side effects of classical analgesics. In support of these concepts, in this article, we describe the functioning of peripheral opioid receptors, migration of opioid-producing immune cells to inflamed tissue, opioid peptide release, and the consequent pain relief. Conclusively, we provide clinical evidence and discuss therapeutic opportunities and challenges associated with immune cell-mediated peripheral opioid analgesia.
Collapse
Affiliation(s)
- Halina Machelska
- Department of Experimental Anesthesiology, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany.
| | - Melih Ö Celik
- Department of Experimental Anesthesiology, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
11
|
Zhou YQ, Liu DQ, Chen SP, Chen N, Sun J, Wang XM, Li DY, Tian YK, Ye DW. PPARγ activation mitigates mechanical allodynia in paclitaxel-induced neuropathic pain via induction of Nrf2/HO-1 signaling pathway. Biomed Pharmacother 2020; 129:110356. [PMID: 32535388 DOI: 10.1016/j.biopha.2020.110356] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Paclitaxel-induced neuropathic pain (PINP) is a dose-limiting side effect and is refractory to widely used analgesic drugs. Previous studies have demonstrated a protective role of peroxisome proliferator-activated receptor gama (PPARγ) in neuropathic pain. However, whether PPARγ activation could alleviate PINP remains to be elucidated. Our previous study has validated the analgesic effect of oltipraz, an nuclear factor erythroid-2 related factor 2 (Nrf2) activator, in a rat model of PINP. In this study, we tested the hypothesis that rosiglitazone, a selective agonist of PPARγ, could attenuate PINP through induction of Nrf2/heme oxygenase-1 (HO-1) signaling pathway. Paclitaxel was injected intraperitoneally on four alternate days to induce neuropathic pain. Paw withdrawal threshold was used to evaluate mechanical allodynia. Western blot and immunofluorescence were used to examine the expression and distribution of PPARγ, Nrf2 and HO-1 in the spinal cord. Our results showed that rosiglitazone attenuated established PINP and delayed the onset of PINP via activation of PPARγ, which were reversed by PPARγ antagonist GW9662. Moreover, rosiglitazone inhibited downregulation of PPARγ in the spinal cord of PINP rats. Furthermore, the analgesic effect of rosiglitazone against PINP was abolished by trigonelline, an Nrf2 inhibitor. Finally, rosiglitazone significantly increased expression of Nrf2 and HO-1 in the spinal cord of PINP rats. Collectively, these results indicated that PPARγ activation might mitigate PINP through activating spinal Nrf2/HO-1 signaling pathway. Our results may provide an alternative option for PINP patients.
Collapse
Affiliation(s)
- Ya-Qun Zhou
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dai-Qiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Ping Chen
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Chen
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Sun
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Mei Wang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Yang Li
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Ke Tian
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Westlund KN, Zhang M. Building and Testing PPARγ Therapeutic ELB00824 with an Improved Therapeutic Window for Neuropathic Pain. Molecules 2020; 25:E1120. [PMID: 32138198 PMCID: PMC7179195 DOI: 10.3390/molecules25051120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 11/20/2022] Open
Abstract
Effective, non-addictive therapeutics for chronic pain remain a critical need. While there are several potential therapeutics that stimulate anti-inflammatory mechanisms to restore homeostasis in the spinal dorsal horn microenvironment, the effectiveness of drugs for neuropathic pain are still inadequate. The convergence of increasing knowledge about the multi-factorial mechanisms underlying neuropathic pain and the mechanisms of drug action from preclinical studies are providing the ability to create pharmaceuticals with better clinical effectiveness. By targeting and activating the peroxisome proliferator-activated receptor gamma subunit (PPARγ), numerous preclinical studies report pleiotropic effects of thiazolidinediones (TDZ) beyond their intended use of increasing insulin, including their anti-inflammatory, renal, cardioprotective, and oncopreventative effects. Several studies find TDZs reduce pain-related behavioral symptoms, including ongoing secondary hypersensitivity driven by central sensitization. Previous studies find increased PPARγ in the spinal cord and brain regions innervated by incoming afferent nerve endings after the induction of neuropathic pain models. PPARγ agonist treatment provides an effective reduction in pain-related behaviors, including anxiety. Data further suggest that improved brain mitochondrial bioenergetics after PPARγ agonist treatment is a key mechanism for reducing hypersensitivity. This review emphasizes two points relevant for the development of better chronic pain therapies. First, employing neuropathic pain models with chronic duration is critical since they can encompass the continuum of molecular and brain circuitry alterations arising over time when pain persists, providing greater relevance to clinical pain syndromes. Assisting in that effort are preclinical models of chronic trigeminal pain syndromes. Secondly, considering the access to nerve and brain neurons and glia across the blood-brain barrier is important. While many therapies have low brain penetrance, a PPARγ agonist with better brain penetrance, ELB00824, has been developed. Purposeful design and recent comparative testing indicate that ELB00824 is extraordinarily efficient and efficacious. ELB00824 provides greatly improved attenuation of pain-related behaviors, including mechanical hypersensitivity, anxiety, and depression in our chronic trigeminal nerve injury models. Physiochemical properties allowing significant brain access and toxicity testing are discussed.
Collapse
Affiliation(s)
- Karin N. Westlund
- Department of Anesthesiology & Critical Care Medicine, MSC10 6000, 2211 Lomas Blvd. NE, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Morgan Zhang
- USA Elixiria Biotech Inc, 200 High Point Drive, Hartsdale, NY 10530, USA;
| |
Collapse
|
13
|
Quintão NLM, Santin JR, Stoeberl LC, Corrêa TP, Melato J, Costa R. Pharmacological Treatment of Chemotherapy-Induced Neuropathic Pain: PPARγ Agonists as a Promising Tool. Front Neurosci 2019; 13:907. [PMID: 31555078 PMCID: PMC6722212 DOI: 10.3389/fnins.2019.00907] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy-induced neuropathic pain (CINP) is one of the most severe side effects of anticancer agents, such as platinum- and taxanes-derived drugs (oxaliplatin, cisplatin, carboplatin and paclitaxel). CINP may even be a factor of interruption of treatment and consequently increasing the risk of death. Besides that, it is important to take into consideration that the incidence of cancer is increasing worldwide, including colorectal, gastric, lung, cervical, ovary and breast cancers, all treated with the aforementioned drugs, justifying the concern of the medical community about the patient’s quality of life. Several physiopathological mechanisms have already been described for CINP, such as changes in axonal transport, mitochondrial damage, increased ion channel activity and inflammation in the central nervous system (CNS). Another less frequent event that may occur after chemotherapy, particularly under oxaliplatin treatment, is the central neurotoxicity leading to disorders such as mental confusion, catatonia, hyporeflexia, etc. To date, no pharmacological therapy has shown satisfactory effect in these cases. In this scenario, duloxetine is the only drug currently in clinical use. Peroxisome proliferator-activated receptors (PPARs) belong to the class of nuclear receptors and are present in several tissues, mainly participating in lipid and glucose metabolism and inflammatory response. There are three PPAR isoforms: α, β/δ and γ. PPARγ, the protagonist of this review, is expressed in adipose tissue, large intestine, spleen and neutrophils. This subtype also plays important role in energy balance, lipid biosynthesis and adipogenesis. The effects of PPARγ agonists, known for their positive activity on type II diabetes mellitus, have been explored and present promising effects in the control of neuropathic pain, including CINP, and also cancer. This review focuses largely on the mechanisms involved in chemotherapy-induced neuropathy and the effects of the activation of PPARγ to treat CINP. It is the aim of this review to help understanding and developing novel CINP therapeutic strategies integrating PPARγ signalling.
Collapse
Affiliation(s)
| | | | | | | | - Jéssica Melato
- School of Heath Science, Universidade do Vale do Itajaí, Itajaí, Brazil
| | - Robson Costa
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
14
|
Momotori N, Jo JI, Tabata Y. Preparation of polymer microspheres capable for pioglitazone release to modify macrophages function. Regen Ther 2019; 11:131-138. [PMID: 31338392 PMCID: PMC6626069 DOI: 10.1016/j.reth.2019.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/12/2019] [Accepted: 06/25/2019] [Indexed: 01/22/2023] Open
Abstract
Introduction Macrophages play an important role in regulating inflammation and tissue regeneration. It is known that anti-inflammatory macrophages play an important role for tissue regeneration. The objective of this study is to modify macrophages phenotypes for anti-inflammatory function by utilizing drug delivery technology. Method In this study, 4 types of poly (L-lactic-co-glycolic acid) (PLGA) microspheres incorporating pioglitazone of an anti-inflammatory modifier (pio-MS) with different sizes were prepared. In vitro release test of pio-MS was performed in phosphate buffered-saline solution (PBS) containing 1 wt% of sodium lauryl sulfate. The arginase activity and the secretion of interleukin (IL)−10 as anti-inflammatory macrophage markers of mouse bone marrow derived-macrophages (BMDM) cultured with the pio-MS were evaluated. Results The sustained release of pioglitazone was observed from all types of pio-MS in vitro. When BMDM were cultured with the pio-MS with an average diameter of 40 μm (pio-MS40), the arginase activity and the secretion of IL-10 increased to a significant extent compared with other pio-MS. Conclusions The pio-MS40 with an diameter of 40 μm had a potential to induce the anti-inflammatory modification of BMDM in this culture system. The sustained release of pioglitazone is promoting to modify the macrophage function. Microspheres incorporating pioglitazone with different sizes were prepared. Sustained release of pioglitazone from the microspheres were observed. The effect of pioglitazone on macrophages was enhanced by the sustained release.
Collapse
Key Words
- BMDM, mouse bone marrow derived-macrophage
- DDW, double-distilled water
- Drug delivery system
- ELISA, enzyme-linked immunosorbent assay
- FBS, fetal bovine serum
- HPLC, high performance liquid chromatography
- IL, interleukin
- IMDM, Iscove's modified Dulbecco's medium
- M-CSF, macrophage colony stimulating factor
- Macrophages
- Microspheres
- PBS, phosphate buffered-saline solution
- PCR, polymerase chain reaction
- PGA, poly(glycolic acid)
- PLA, poly(l-lactic acid)
- PLGA
- PLGA, poly(L-lactic-co-glycolic acid)
- PPARγ, peroxisome proliferator-activated receptor γ
- Pioglitazone
- Poly(L-lactic-co-glycolic acid)
- RS, resulting solution
- SD, standard deviation
- SEM, scanning electron microscopy
- TNF, tumor necrosis factor
- UV, ultra violet
- iNOS, inducible nitric oxide synthase
- pio-MS, PLGA microspheres incorporating pioglitazone
Collapse
Affiliation(s)
- Naoki Momotori
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
15
|
Gazerani P. Identification of novel analgesics through a drug repurposing strategy. Pain Manag 2019; 9:399-415. [DOI: 10.2217/pmt-2018-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The identification of new indications for approved or failed drugs is a process called drug repositioning or drug repurposing. The motivation includes overcoming the productivity gap that exists in drug development, which is a high-cost–high-risk process. Repositioning also includes rescuing drugs that have safely entered the market but have failed to demonstrate sufficient efficiency for the initial clinical indication. Considering the high prevalence of chronic pain, the lack of sufficient efficacy and the safety issues of current analgesics, repositioning seems to be an attractive approach. This review presents example of drugs that already have been repositioned and highlights new technologies that are available for the identification of additional compounds to stimulate the curiosity of readers for further exploration.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine, Department of Health Science & Technology, Aalborg University, Frederik Bajers Vej 3 B, 9220 Aalborg East, Denmark
| |
Collapse
|
16
|
Effects of Dual Peroxisome Proliferator-Activated Receptors α and γ Activation in Two Rat Models of Neuropathic Pain. PPAR Res 2019; 2019:2630232. [PMID: 31139213 PMCID: PMC6500665 DOI: 10.1155/2019/2630232] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/02/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Neuropathic pain is a growing healthcare problem causing a global burden. Currently used analgesics such as opioids are associated with adverse effects; urging the need for safer alternatives. Here we aimed to investigate the potential analgesic effects of tesaglitazar; dual peroxisome proliferator-activated receptors α and γ (PPARα and γ) agonist in rat models of neuropathic pain. This study also aimed to investigate the modulation of the transient receptor potential vanilloid 1 (TRPV1) receptor activity by tesaglitazar which could provide a potential mechanism that underlie tesaglitazar antinociceptive effects. Von Frey filaments were used to determine the paw withdrawal threshold (PWT) in adult male Sprague Dawley rats (180-250g) following i.p. injection of streptozotocin (STZ) or cisplatin, which were used as models of neuropathic pain. Antinociceptive effects of tesaglitazar were determined 6 hours after drug administration. Cobalt influx assays in cultured dorsal root ganglia (DRG) neurons were used to study the effects of tesaglitazar preincubation on capsaicin-evoked cobalt influx. Both cisplatin and STZ produced a significant decrease in PWT. The higher dose of tesaglitazar (20μg/kg) significantly restored PWT in both neuropathic pain models (P<0.05). 10μM capsaicin produced a robust cobalt response in DRG neurons. Preincubation of DRG neurones with tesaglitazar 6 hours prior to stimulation with capsaicin significantly reduce capsaicin-evoked cobalt responses in a PPARα and PPARγ dependent fashion (P<0.05). In conclusion, tesaglitazar produced significant analgesic effects in STZ and cisplatin-induced neuropathy, possibly by modulating TRPV1 receptor activity. This may be of potential benefit in clinical practice dealing with peripheral neuropathy.
Collapse
|
17
|
Okine BN, Gaspar JC, Finn DP. PPARs and pain. Br J Pharmacol 2018; 176:1421-1442. [PMID: 29679493 DOI: 10.1111/bph.14339] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/19/2018] [Accepted: 03/26/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic pain is a common cause of disability worldwide and remains a global health and socio-economic challenge. Current analgesics are either ineffective in a significant proportion of patients with chronic pain or associated with significant adverse side effects. The PPARs, a family of nuclear hormone transcription factors, have emerged as important modulators of pain in preclinical studies and therefore a potential therapeutic target for the treatment of pain. Modulation of nociceptive processing by PPARs is likely to involve both transcription-dependent and transcription-independent mechanisms. This review presents a comprehensive overview of preclinical studies investigating the contribution of PPAR signalling to nociceptive processing in animal models of inflammatory and neuropathic pain. We examine current evidence from anatomical, molecular and pharmacological studies demonstrating a role for PPARs in pain control. We also discuss the limited evidence available from relevant clinical studies and identify areas that warrant further research. LINKED ARTICLES: This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.
Collapse
Affiliation(s)
- Bright N Okine
- Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland.,Centre for Pain Research, NCBES, National University of Ireland Galway, Galway, Ireland
| | - Jessica C Gaspar
- Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland.,Centre for Pain Research, NCBES, National University of Ireland Galway, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, National University of Ireland Galway, Galway, Ireland.,Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland.,Centre for Pain Research, NCBES, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
18
|
Abstract
Neuropathic pain represents a significant and mounting burden on patients and society at large. Management of neuropathic pain, however, is both intricate and challenging, exacerbated by the limited quantity and quality of clinically available treatments. On this stage, dysfunctional voltage-gated ion channels, especially the presynaptic N-type voltage-gated calcium channel (VGCC) (Cav2.2) and the tetrodotoxin-sensitive voltage-gated sodium channel (VGSC) (Nav1.7), underlie the pathophysiology of neuropathic pain and serve as high profile therapeutic targets. Indirect regulation of these channels holds promise for the treatment of neuropathic pain. In this review, we focus on collapsin response mediator protein 2 (CRMP2), a protein with emergent roles in voltage-gated ion channel trafficking and discuss the therapeutic potential of targetting this protein.
Collapse
|
19
|
Mansouri MT, Naghizadeh B, Ghorbanzadeh B, Rajabi H, Pashmforoush M. Pharmacological evidence for systemic and peripheral antinociceptive activities of pioglitazone in the rat formalin test: Role of PPARγ and nitric oxide. Eur J Pharmacol 2017; 805:84-92. [DOI: 10.1016/j.ejphar.2017.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 02/11/2017] [Accepted: 03/08/2017] [Indexed: 12/19/2022]
|
20
|
Pannell M, Labuz D, Celik MÖ, Keye J, Batra A, Siegmund B, Machelska H. Adoptive transfer of M2 macrophages reduces neuropathic pain via opioid peptides. J Neuroinflammation 2016; 13:262. [PMID: 27717401 PMCID: PMC5055715 DOI: 10.1186/s12974-016-0735-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022] Open
Abstract
Background During the inflammation which occurs following nerve damage, macrophages are recruited to the site of injury. Phenotypic diversity is a hallmark of the macrophage lineage and includes pro-inflammatory M1 and anti-inflammatory M2 populations. Our aim in this study was to investigate the ability of polarized M0, M1, and M2 macrophages to secrete opioid peptides and to examine their relative contribution to the modulation of neuropathic pain. Methods Mouse bone marrow-derived cells were cultured as unstimulated M0 macrophages or were stimulated into an M1 phenotype using lipopolysaccharide and interferon-γ or into an M2 phenotype using interleukin-4. The macrophage phenotypes were verified using flow cytometry for surface marker analysis and cytokine bead array for cytokine profile assessment. Opioid peptide levels were measured by radioimmunoassay and enzyme immunoassay. As a model of neuropathic pain, a chronic constriction injury (CCI) of the sciatic nerve was employed. Polarized M0, M1, and M2 macrophages (5 × 105 cells) were injected perineurally twice, on days 14 and 15 following CCI or sham surgery. Mechanical and heat sensitivity were measured using the von Frey and Hargreaves tests, respectively. To track the injected macrophages, we also transferred fluorescently stained polarized cells and analyzed the surface marker profile of endogenous and injected cells in the nerves ex vivo. Results Compared to M0 and M1 cells, M2 macrophages contained and released higher amounts of opioid peptides, including Met-enkephalin, dynorphin A (1–17), and β-endorphin. M2 cells transferred perineurally at the nerve injury site reduced mechanical, but not heat hypersensitivity following the second injection. The analgesic effect was reversed by the perineurally applied opioid receptor antagonist naloxone methiodide. M2 cells did not affect sensitivity following sham surgery. Neither M0 nor M1 cells altered mechanical and heat sensitivity in CCI or sham-operated animals. Tracing the fluorescently labeled M0, M1, and M2 cells ex vivo showed that they remained in the nerve and preserved their phenotype. Conclusions Perineural transplantation of M2 macrophages resulted in opioid-mediated amelioration of neuropathy-induced mechanical hypersensitivity, while M1 macrophages did not exacerbate pain. Therefore, rather than focusing on macrophage-induced pain generation, promoting opioid-mediated M2 actions may be more relevant for pain control.
Collapse
Affiliation(s)
- Maria Pannell
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Dominika Labuz
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Melih Ö Celik
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Jacqueline Keye
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Arvind Batra
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Britta Siegmund
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Halina Machelska
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
21
|
Jia H, Xu S, Liu Q, Liu J, Xu J, Li W, Jin Y, Ji Q. Effect of pioglitazone on neuropathic pain and spinal expression of TLR-4 and cytokines. Exp Ther Med 2016; 12:2644-2650. [PMID: 27698768 DOI: 10.3892/etm.2016.3643] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 01/08/2015] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanisms underlying neuropathic pain have yet to be elucidated. The present study aimed to examine the modulation of neuroimmune activation in the spinal cord by the synthetic peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, pioglitazone (Pio), in a rat model of neuropathic pain induced by chronic constriction injury (CCI). Rats were randomly assigned into four groups: Sham surgery with vehicle, chronic constriction injury with vehicle or Pio (10 mg/kg), and chronic constriction injury with Pio and a PPAR-γ antagonist GW9662 (2 mg/kg). Pio or vehicle was administered 1 h prior to the surgery and continued daily until day 7 post-surgery. Paw pressure threshold was measured prior to surgery and on days 0, 1, 3 and 7 post-surgery. Microglia activation markers macrophage antigen complex-1, the mRNA expression levels of tumor necrosis factor α and interleukin-1β, and the mRNA expression levels of toll like receptor (TLR-4) in the lumbar spinal cord were determined. Administration of Pio resulted in the prominent attenuation of mechanical hyperalgesia. In addition, Pio was able to significantly inhibit neuroimmune activation characterized by glial activation, the production of cytokines and expression levels of TLR-4. Concurrent administration of a PPAR-γ antagonist, GW9662, reversed the effects of Pio. The antihyperalgesic effect of administration of Pio in rats receiving CCI may, in part, be attributed to the inhibition of neuroimmune activation associated with the sustaining of neuropathic pain.
Collapse
Affiliation(s)
- Hongbin Jia
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Shuangshuang Xu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jian Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Jianguo Xu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Weiyan Li
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yi Jin
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qing Ji
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
22
|
Macedo CG, Napimoga MH, Rocha-Neto LM, Abdalla HB, Clemente-Napimoga JT. The role of endogenous opioid peptides in the antinociceptive effect of 15-deoxy(Δ12,14)-prostaglandin J2 in the temporomandibular joint. Prostaglandins Leukot Essent Fatty Acids 2016; 110:27-34. [PMID: 27255640 DOI: 10.1016/j.plefa.2016.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
Abstract
We have previously demonstrated that peripheral administration of 15d-PGJ2 in the Temporomandibular joint (TMJ) of rats can prevent nociceptor sensitization, mediated by peroxisome proliferator activated receptor-γ (PPAR-γ), and κ- and δ- opioid receptors. However, the mechanism that underlies the signaling of PPAR-γ (upon activation by 15d-PGJ2) to induce antinociception, and how the opioid receptors are activated via 15d-PGJ2 are not fully understood. This study demonstrates that peripheral antinociceptive effect of 15d-PGJ2 is mediated by PPAR-γ expressed in the inflammatory cells of TMJ tissues. Once activated by 15d-PGJ2, PPAR-γ induces the release of β-endorphin and dynorphin, which activates κ- and δ-opioid receptors in primary sensory neurons to induce the antinociceptive effect.
Collapse
Affiliation(s)
- C G Macedo
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Av. Limeira, 901, Piracicaba, SP 13414-903, Brazil
| | - M H Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Rua José Rocha Junqueira, 13 - Campinas, SP 13045-755, Brazil
| | - L M Rocha-Neto
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Av. Limeira, 901, Piracicaba, SP 13414-903, Brazil
| | - H B Abdalla
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Av. Limeira, 901, Piracicaba, SP 13414-903, Brazil
| | - J T Clemente-Napimoga
- Department of Physiological Sciences, Laboratory of Orofacial Pain, Piracicaba Dental School, University of Campinas - UNICAMP, Av. Limeira, 901, Piracicaba, SP 13414-903, Brazil.
| |
Collapse
|
23
|
Abstract
Introduction There is growing interest in whether social media can capture patient-generated information relevant for medicines safety surveillance that cannot be found in traditional sources. Objective The aim of this study was to evaluate the potential contribution of mining social media networks for medicines safety surveillance using the following associations as case studies: (1) rosiglitazone and cardiovascular events (i.e. stroke and myocardial infarction); and (2) human papilloma virus (HPV) vaccine and infertility. Methods We collected publicly accessible, English-language posts on Facebook, Google+, and Twitter until September 2014. Data were queried for co-occurrence of keywords related to the drug/vaccine and event of interest within a post. Messages were analysed with respect to geographical distribution, context, linking to other web content, and author’s assertion regarding the supposed association. Results A total of 2537 posts related to rosiglitazone/cardiovascular events and 2236 posts related to HPV vaccine/infertility were retrieved, with the majority of posts representing data from Twitter (98 and 85 %, respectively) and originating from users in the US. Approximately 21 % of rosiglitazone-related posts and 84 % of HPV vaccine-related posts referenced other web pages, mostly news items, law firms’ websites, or blogs. Assertion analysis predominantly showed affirmation of the association of rosiglitazone/cardiovascular events (72 %; n = 1821) and of HPV vaccine/infertility (79 %; n = 1758). Only ten posts described personal accounts of rosiglitazone/cardiovascular adverse event experiences, and nine posts described HPV vaccine problems related to infertility. Conclusions Publicly available data from the considered social media networks were sparse and largely untrackable for the purpose of providing early clues of safety concerns regarding the prespecified case studies. Further research investigating other case studies and exploring other social media platforms are necessary to further characterise the usefulness of social media for safety surveillance. Electronic supplementary material The online version of this article (doi:10.1007/s40264-015-0333-5) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Griggs RB, Donahue RR, Adkins BG, Anderson KL, Thibault O, Taylor BK. Pioglitazone Inhibits the Development of Hyperalgesia and Sensitization of Spinal Nociresponsive Neurons in Type 2 Diabetes. THE JOURNAL OF PAIN 2016; 17:359-73. [PMID: 26687453 PMCID: PMC4791042 DOI: 10.1016/j.jpain.2015.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/13/2015] [Accepted: 11/25/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED Thiazolidinedione drugs (TZDs) such as pioglitazone are approved by the U.S. Food and Drug Administration for the treatment of insulin resistance in type 2 diabetes. However, whether TZDs reduce painful diabetic neuropathy (PDN) remains unknown. Therefore, we tested the hypothesis that chronic administration of pioglitazone would reduce PDN in Zucker Diabetic Fatty (ZDF(fa/fa) [ZDF]) rats. Compared with Zucker Lean (ZL(fa/+)) controls, ZDF rats developed: (1) increased blood glucose, hemoglobin A1c, methylglyoxal, and insulin levels; (2) mechanical and thermal hyperalgesia in the hind paw; (3) increased avoidance of noxious mechanical probes in a mechanical conflict avoidance behavioral assay, to our knowledge, the first report of a measure of affective-motivational pain-like behavior in ZDF rats; and (4) exaggerated lumbar dorsal horn immunohistochemical expression of pressure-evoked phosphorylated extracellular signal-regulated kinase. Seven weeks of pioglitazone (30 mg/kg/d in food) reduced blood glucose, hemoglobin A1c, hyperalgesia, and phosphorylated extracellular signal-regulated kinase expression in ZDF. To our knowledge, this is the first report to reveal hyperalgesia and spinal sensitization in the same ZDF animals, both evoked by a noxious mechanical stimulus that reflects pressure pain frequently associated with clinical PDN. Because pioglitazone provides the combined benefit of reducing hyperglycemia, hyperalgesia, and central sensitization, we suggest that TZDs represent an attractive pharmacotherapy in patients with type 2 diabetes-associated pain. PERSPECTIVE To our knowledge, this is the first preclinical report to show that: (1) ZDF rats exhibit hyperalgesia and affective-motivational pain concurrent with central sensitization; and (2) pioglitazone reduces hyperalgesia and spinal sensitization to noxious mechanical stimulation within the same subjects. Further studies are needed to determine the anti-PDN effect of TZDs in humans.
Collapse
Affiliation(s)
- Ryan B Griggs
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Renee R Donahue
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Braxton G Adkins
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Katie L Anderson
- Department of Pharmacology and Nutritional Science, College of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Science, College of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Bradley K Taylor
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, Lexington, Kentucky.
| |
Collapse
|
25
|
Martins DF, Prado MRB, Daruge-Neto E, Batisti AP, Emer AA, Mazzardo-Martins L, Santos ARS, Piovezan AP. Caffeine prevents antihyperalgesic effect of gabapentin in an animal model of CRPS-I: evidence for the involvement of spinal adenosine A1
receptor. J Peripher Nerv Syst 2015; 20:403-9. [DOI: 10.1111/jns.12149] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Daniel F. Martins
- Post-Graduate Program of Health Sciences; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Experimental Neuroscience Laboratory; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Undergraduate Course of Medicine; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
| | - Marcos R. B. Prado
- Experimental Neuroscience Laboratory; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Undergraduate Course of Medicine; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
| | - Eduardo Daruge-Neto
- Experimental Neuroscience Laboratory; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Undergraduate Course of Medicine; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
| | - Ana P. Batisti
- Experimental Neuroscience Laboratory; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
| | - Aline A. Emer
- Post-Graduate Program of Health Sciences; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Experimental Neuroscience Laboratory; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
| | - Leidiane Mazzardo-Martins
- Department of Morphological Sciences; Federal University of Santa Catarina; Florianópolis Santa Catarina Brazil
| | - Adair R. S. Santos
- Neurobiology Laboratory of Pain and Inflammation, Department of Physiological Sciences, Center of Biological Sciences; Federal University of Santa Catarina; Florianópolis Santa Catarina Brazil
| | - Anna P. Piovezan
- Post-Graduate Program of Health Sciences; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Experimental Neuroscience Laboratory; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
- Undergraduate Course of Medicine; University of Southern Santa Catarina; Palhoça Santa Catarina Brazil
| |
Collapse
|
26
|
Abstract
Repeated administration of peroxisome proliferator-activated receptor gamma (PPARγ) agonists reduces neuropathic pain-like behavior and associated changes in glial activation in the spinal cord dorsal horn. As PPARγ is a nuclear receptor, sustained changes in gene expression are widely believed to be the mechanism of pain reduction. However, we recently reported that a single intrathecal (i.t.) injection of pioglitazone, a PPARγ agonist, reduced hyperalgesia within 30 minutes, a time frame that is typically less than that required for genomic mechanisms. To determine the very rapid antihyperalgesic actions of PPARγ activation, we administered pioglitazone to rats with spared nerve injury and evaluated hyperalgesia. Pioglitazone inhibited hyperalgesia within 5 minutes of injection, consistent with a nongenomic mechanism. Systemic or i.t. administration of GW9662, a PPARγ antagonist, inhibited the antihyperalgesic actions of intraperitoneal or i.t. pioglitazone, suggesting a spinal PPARγ-dependent mechanism. To further address the contribution of nongenomic mechanisms, we blocked new protein synthesis in the spinal cord with anisomycin. When coadministered intrathecally, anisomycin did not change pioglitazone antihyperalgesia at an early 7.5-minute time point, further supporting a rapid nongenomic mechanism. At later time points, anisomycin reduced pioglitazone antihyperalgesia, suggesting delayed recruitment of genomic mechanisms. Pioglitazone reduction of spared nerve injury-induced increases in GFAP expression occurred more rapidly than expected, within 60 minutes. We are the first to show that activation of spinal PPARγ rapidly reduces neuropathic pain independent of canonical genomic activity. We conclude that acute pioglitazone inhibits neuropathic pain in part by reducing astrocyte activation and through both genomic and nongenomic PPARγ mechanisms.
Collapse
|
27
|
Adiponectin-Mediated Analgesia and Anti-Inflammatory Effects in Rat. PLoS One 2015; 10:e0136819. [PMID: 26352808 PMCID: PMC4564279 DOI: 10.1371/journal.pone.0136819] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/10/2015] [Indexed: 02/02/2023] Open
Abstract
The adipose tissue-derived protein, adiponectin, has significant anti-inflammatory properties in a variety of disease conditions. Recent evidence that adiponectin and its receptors (AdipoR1 and AdipoR2) are expressed in central nervous system, suggests that it may also have a central modulatory role in pain and inflammation. This study set out to investigate the effects of exogenously applied recombinant adiponectin (via intrathecal and intraplantar routes; 10–5000 ng) on the development of peripheral inflammation (paw oedema) and pain hypersensitivity in the rat carrageenan model of inflammation. Expression of adiponectin, AdipoR1 and AdipoR2 mRNA and protein was characterised in dorsal spinal cord using real-time polymerase chain reaction (PCR) and Western blotting. AdipoR1 and AdipoR2 mRNA and protein were found to be constitutively expressed in dorsal spinal cord, but no change in mRNA expression levels was detected in response to carrageenan-induced inflammation. Adiponectin mRNA, but not protein, was detected in dorsal spinal cord, although levels were very low. Intrathecal administration of adiponectin, both pre- and 3 hours post-carrageenan, significantly attenuated thermal hyperalgesia and mechanical hypersensitivity. Intrathecal administration of adiponectin post-carrageenan also reduced peripheral inflammation. Intraplantar administration of adiponectin pre-carrageenan dose-dependently reduced thermal hyperalgesia but had no effect on mechanical hypersensitivity and peripheral inflammation. These results show that adiponectin functions both peripherally and centrally at the spinal cord level, likely through activation of AdipoRs to modulate pain and peripheral inflammation. These data suggest that adiponectin receptors may be a novel therapeutic target for pain modulation.
Collapse
|
28
|
de Guglielmo G, Kallupi M, Scuppa G, Stopponi S, Demopulos G, Gaitanaris G, Ciccocioppo R. Analgesic tolerance to morphine is regulated by PPARγ. Br J Pharmacol 2015; 171:5407-16. [PMID: 25048682 DOI: 10.1111/bph.12851] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 06/23/2014] [Accepted: 07/14/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Opioid drugs are potent analgesics. However, their chronic use leads to the rapid development of tolerance to their analgesic effects and subsequent increase of significant side effects, including drug dependence and addiction. Here, we investigated the role of PPARγ in the development of analgesic tolerance to morphine in mice. EXPERIMENTAL APPROACH We monitored analgesia on alternate days using the tail immersion test. KEY RESULTS Daily administration of morphine (30 mg·kg(-1) , bid) resulted in the rapid development of tolerance to thermal analgesia. Co-administration of pioglitazone (10 and 30 mg·kg(-1) , bid) significantly attenuated the development and expression of tolerance. However, pretreatment with GW-9662 (5 mg·kg(-1) , bid), a selective PPARγ antagonist, completely abolished this effect. Injection of GW-9662 and a lower dose of morphine (15 mg·kg(-1) , bid) accelerated the development of tolerance to its antinociceptive effect. Subsequently, we found that conditional neuronal PPARγ knockout (KO) mice develop a more rapid and pronounced tolerance to morphine antinociception compared with wild-type (WT) controls. Moreover, in PPARγ KO mice, pioglitazone was no longer able to prevent the development of morphine tolerance. CONCLUSIONS AND IMPLICATIONS Overall, our results demonstrate that PPARγ plays a tonic role in the modulation of morphine tolerance, and its pharmacological activation may help to reduce its development. These findings provide new information about the role of neuronal PPARγ and suggest that combining PPARγ agonists with opioid analgesics may reduce the development of tolerance and possibly attenuate the potential for opioid abuse.
Collapse
|
29
|
Pottabathini R, Kumar A, Bhatnagar A, Garg S, Ekavali E. Ameliorative potential of pioglitazone and ceftriaxone alone and in combination in rat model of neuropathic pain: Targeting PPARγ and GLT-1 pathways. Pharmacol Rep 2015; 68:85-94. [PMID: 26721358 DOI: 10.1016/j.pharep.2015.06.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/15/2015] [Accepted: 06/24/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND The relation between glutamate homeostasis and PPAR gamma has got tremendous importance in nerve trauma and pain. Present study has been designed to elucidate the interaction between the GLT-1 activator (ceftriaxone) and PPAR gamma agonist (pioglitazone) in the spinal nerve ligation induced neuropathic pain. METHODS Male SD rats were subjected to spinal nerve ligation to induce neuropathic pain. Pioglitazone, ceftriaxone and their combination treatments were given for 28 days. Various behavioral, biochemical, neuroinflammatory and apoptotic mediators were assessed subsequently. RESULTS In the present study, ligation of L5 and L6 spinal nerves resulted in marked hyperalgesia and allodynia to different mechanical and thermal stimuli. In addition there is marked increase in oxidative-nitrosative stress parameters, inflammatory and apoptotic markers in spinal cord of spinal nerve ligated rats. Treatment with pioglitazone and ceftriaxone significantly prevented these behavioral, biochemical, mitochondrial and cellular alterations in rats. Further, combination of pioglitazone (10mg/kg, ip) with ceftriaxone (100mg/kg, ip) significantly potentiated the protective effects as compared to their effects per se. CONCLUSION Based on these results we propose that possible interplay between the neuroprotective effects of pioglitazone and ceftriaxone exists in suppressing the behavioral, biochemical, mitochondrial, neuroinflammatory and apoptotic cascades in spinal nerve ligation induced neuropathic pain in rats.
Collapse
Affiliation(s)
- Raghavender Pottabathini
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India.
| | | | - Sukant Garg
- Department of Pathology, Dr. Harvansh Singh Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, India
| | - E Ekavali
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, India
| |
Collapse
|
30
|
Sato K, Sakai S, Kishi K, Tabata Y. Controlled release of pioglitazone from biodegradable hydrogels to modify macrophages phenotype. Inflamm Regen 2015. [DOI: 10.2492/inflammregen.35.086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Keisuke Sato
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shigeki Sakai
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Freitag CM, Miller RJ. Peroxisome proliferator-activated receptor agonists modulate neuropathic pain: a link to chemokines? Front Cell Neurosci 2014; 8:238. [PMID: 25191225 PMCID: PMC4138931 DOI: 10.3389/fncel.2014.00238] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/28/2014] [Indexed: 11/29/2022] Open
Abstract
Chronic pain presents a widespread and intractable medical problem. While numerous pharmaceuticals are used to treat chronic pain, drugs that are safe for extended use and highly effective at treating the most severe pain do not yet exist. Chronic pain resulting from nervous system injury (neuropathic pain) is common in conditions ranging from multiple sclerosis to HIV-1 infection to type II diabetes. Inflammation caused by neuropathy is believed to contribute to the generation and maintenance of neuropathic pain. Chemokines are key inflammatory mediators, several of which (MCP-1, RANTES, MIP-1α, fractalkine, SDF-1 among others) have been linked to chronic, neuropathic pain in both human conditions and animal models. The important roles chemokines play in inflammation and pain make them an attractive therapeutic target. Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptors known for their roles in metabolism. Recent research has revealed that PPARs also play a role in inflammatory gene repression. PPAR agonists have wide-ranging effects including inhibition of chemokine expression and pain behavior reduction in animal models. Experimental evidence suggests a connection between the pain ameliorating effects of PPAR agonists and suppression of inflammatory gene expression, including chemokines. In early clinical research, one PPARα agonist, palmitoylethanolamide (PEA), shows promise in relieving chronic pain. If this link can be better established, PPAR agonists may represent a new drug therapy for neuropathic pain.
Collapse
Affiliation(s)
- Caroline M Freitag
- Department of Molecular Pharmacology and Biological Chemistry, Richard J. Miller Laboratory, Northwestern University Chicago, IL, USA
| | - Richard J Miller
- Department of Molecular Pharmacology and Biological Chemistry, Richard J. Miller Laboratory, Northwestern University Chicago, IL, USA
| |
Collapse
|
32
|
Abstract
The number of approved new molecular entity drugs has been decreasing as the pharmaceutical company investment in research and development is increasing. As we face this painful crisis, called an innovation gap, there is increasing awareness that development of new uses of existing drugs may be a powerful tool to help overcome this obstacle because it takes too long, costs too much and can be risky to release drugs developed de novo. Consequently, drug repositioning is emerging in different therapeutic areas, including the pain research area. Worldwide, pain is the main reason for seeking healthcare, and pain relief represents an unmet global clinical need. Therefore, development of analgesics with better efficacy, safety and cost effectiveness is of paramount importance. Despite the remarkable advancement in research on cellular and molecular mechanisms underlying pain pathophysiology over the past three decades, target-based therapeutic opportunities have not been pursued to the same extent. Phenotypic screening remains a more powerful tool for drug development than target-based screening so far. It sounds somewhat heretical, but some multi-action drugs, rather than very selective ones, have been developed intentionally. In the present review, we first critically discuss the utility of drug repositioning for analgesic drug development and then show examples of 'old' drugs that have been successfully repositioned or that are under investigation for their analgesic actions. We conclude that drug repositioning should be more strongly encouraged to help build a bridge between basic research and pain relief worldwide.
Collapse
Affiliation(s)
- Leandro Francisco Silva Bastos
- Laboratório de Imunofarmacologia, sala O4-202, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos 6627, Pampulha, Belo Horizonte, CEP 31.270-901, Brazil,
| | | |
Collapse
|
33
|
Abstract
Traumatic injury or disease of the spinal cord and brain elicits multiple cellular and biochemical reactions that together cause or are associated with neuropathology. Specifically, injury or disease elicits acute infiltration and activation of immune cells, death of neurons and glia, mitochondrial dysfunction, and the secretion of substrates that inhibit axon regeneration. In some diseases, inflammation is chronic or non-resolving. Ligands that target PPARs (peroxisome proliferator-activated receptors), a group of ligand-activated transcription factors, are promising therapeutics for neurologic disease and CNS injury because their activation affects many, if not all, of these interrelated pathologic mechanisms. PPAR activation can simultaneously weaken or reprogram the immune response, stimulate metabolic and mitochondrial function, promote axon growth and induce progenitor cells to differentiate into myelinating oligodendrocytes. PPAR activation has beneficial effects in many pre-clinical models of neurodegenerative diseases and CNS injury; however, the mechanisms through which PPARs exert these effects have yet to be fully elucidated. In this review we discuss current literature supporting the role of PPAR activation as a therapeutic target for treating traumatic injury and degenerative diseases of the CNS.
Collapse
|
34
|
Tai LW, Hung VKL, Mei W, Qiu Q, Chung SK, Cheung CW. Effects of repeated central administration of endothelin type A receptor antagonist on the development of neuropathic pain in rats. BIOMED RESEARCH INTERNATIONAL 2013; 2013:529871. [PMID: 24073407 PMCID: PMC3773389 DOI: 10.1155/2013/529871] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/13/2013] [Accepted: 07/26/2013] [Indexed: 11/18/2022]
Abstract
Endothelin-1 (ET-1) predominates in the endothelin family effectively in vascular tone control, mitogenesis, and neuromodulation. Its receptors are widespread in the central nervous system (CNS) associated with endogenous pain control, suggesting an important role of ET-1 in central pain processing. This study aimed to evaluate the effect of central ET-1 on the development of neuropathic pain behaviour by repeated intrathecal administration of endothelin type A receptor (ETAR) antagonist (BQ-123) in a sciatic nerve ligation (SNL) animal model. BQ-123 was administered intrathecally to rats at dosages 15 μg, 20 μg, 25 μg, and 30 μg, daily for 3 days. Mechanical allodynia was assessed daily 30 minutes before/after injection, 1 hour after injection of BQ-123 from post-SNL day 4 to day 6, and once on day 7 (without BQ-123 administration) before rats were sacrificed. Increasing trends of mechanical threshold were observed, and they reached significance at all dosages on post-SNL day 7 (P < 0.05 at dosage 15 μg and P < 0.001 at dosages 20 μg, 25 μg, and 30 μg) in comparison to control group. BQ-123 at dosage 30 μg showed the most stable and significant mechanical threshold rise. Repeated central administration of BQ-123 alleviated mechanical allodynia after SNL. Our results provide insight into the therapeutic strategies, including timing, against neuropathic pain development with ETAR antagonist.
Collapse
Affiliation(s)
- Lydia W. Tai
- Department of Anaesthesiology, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
| | - Victor K. L. Hung
- Department of Anaesthesiology, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
| | - Wei Mei
- Department of Anaesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qiu Qiu
- Department of Anaesthesiology, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Department of Anaesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Sookja K. Chung
- Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Department of Anatomy, The University of Hong Kong, 1/F, Laboratory Block, Faculty of Medicine Building, 21 Sassoon Road, Hong Kong
- Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, 2/F, William MW Mong Block, Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong
| | - C. W. Cheung
- Department of Anaesthesiology, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Queen Mary Hospital, Room 424, 4/F, Block K, 102 Pokfulam, Pokfulam, Hong Kong
- Research Center of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, 2/F, William MW Mong Block, Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong
| |
Collapse
|
35
|
Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuates inflammatory pain through the induction of heme oxygenase-1 in macrophages. Pain 2013; 154:1402-12. [PMID: 23707273 DOI: 10.1016/j.pain.2013.04.039] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 04/10/2013] [Accepted: 04/17/2013] [Indexed: 11/23/2022]
Abstract
Macrophage infiltration to inflammatory sites promotes tissue repair and may be involved in pain hypersensitivity. Peroxisome proliferator-activated receptor (PPAR)γ signaling is known to regulate polarity of macrophages, which are often referred to as proinflammatory (M1) and antiinflammatory (M2) macrophages. We recently showed that the PPARγ agonist rosiglitazone ameliorated the development of postincisional hyperalgesia by increasing the influx of M2 macrophages to inflamed sites. It has been suggested that heme oxygenase (HO)-1, upregulated by PPARγ signaling, promotes differentiation of macrophages to M2 phenotype. In this study, we investigated how rosiglitazone alters pain hypersensitivity by a PPARγHO-1-dependent mechanism during the course of inflammation induced by complete Freund's adjuvant. Local administration of rosiglitazone alleviated mechanical hyperalgesia, with increased gene induction of HO-1. Phenotype switching of infiltrated macrophages to M2 by rosiglitazone was reversed by an HO-1 inhibitor, tin protoporphyrin, at the inflamed sites. Direct stimulation of peritoneal macrophages with rosiglitazone also increased HO-1 induction in the presence of lipopolysaccharide/interferon-γ. Moreover, rosiglitazone increased gene induction of endogenous opioid proenkephalin, both at inflamed sites and in isolated macrophages. Administration of naloxone blocked the analgesic effects of rosiglitazone. We speculate that rosiglitazone alleviated the development of inflammatory pain, possibly through regulating the M1/M2 balance at the inflamed site by a PPARγ/HO-1-dependent mechanism. PPARγ signaling in macrophages may be a potential therapeutic target for the treatment of acute pain development.
Collapse
|
36
|
Ju W, Li Q, Wilson SM, Brittain JM, Meroueh L, Khanna R. SUMOylation alters CRMP2 regulation of calcium influx in sensory neurons. Channels (Austin) 2013; 7:153-9. [PMID: 23510938 DOI: 10.4161/chan.24224] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The axon/dendrite specification collapsin response mediator protein 2 (CRMP2) bidirectionally modulates N-type voltage-gated Ca ( 2+) channels (CaV2.2). Here we demonstrate that small ubiquitin-like modifier (SUMO) protein modifies CRMP2 via the SUMO E2-conjugating enzyme Ubc9 in vivo. Removal of a SUMO conjugation site KMD in CRMP2 (K374A/M375A/D376A; CRMP2AAA) resulted in loss of SUMOylated CRMP2 without compromising neurite branching, a canonical hallmark of CRMP2 function. Increasing SUMOylation levels correlated inversely with calcium influx in sensory neurons. CRMP2 deSUMOylation by SUMO proteases SENP1 and SENP2 normalized calcium influx to those in the CRMP2AAA mutant. Thus, our results identify a novel role for SUMO modification in CRMP2/CaV2.2 signaling pathway.
Collapse
Affiliation(s)
- Weina Ju
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
37
|
Morgenweck J, Griggs RB, Donahue RR, Zadina JE, Taylor BK. PPARγ activation blocks development and reduces established neuropathic pain in rats. Neuropharmacology 2013; 70:236-46. [PMID: 23415633 DOI: 10.1016/j.neuropharm.2013.01.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 12/18/2012] [Accepted: 01/16/2013] [Indexed: 12/30/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a new pharmacotherapeutic target for chronic pain. When oral (3-30 mg/kg/day in chow for 7 wk) or twice-daily intraperitoneal (1-10 mg/kg/day for 2 wk) administration began before spared nerve injury (SNI), pioglitazone, a PPARγ agonist, dose-dependently prevented multiple behavioral signs of somatosensory hypersensitivity. The highest dose of intraperitoneal pioglitazone did not produce ataxia or reductions in transient mechanical and heat nociception, indicating that inhibitory effects on hypersensitivity were not secondary to adverse drug-induced behaviors or antinociception. Inhibitory effects on hypersensitivity persisted at least one week beyond cessation of pioglitazone administration, suggestive of long-lasting effects on gene expression. Blockade of PPARγ with GW9662, an irreversible and selective PPARγ antagonist, dose-dependently reduced the inhibitory effect of pioglitazone on hypersensitivity, indicating a PPARγ-dependent action. Remarkably, a single preemptive injection of pioglitazone 15 min before SNI attenuated hypersensitivity for at least 2 weeks; this was enhanced with a second injection delivered 12 h after SNI. Pioglitazone injections beginning after SNI also reduced hypersensitivity, albeit to a lesser degree than preemptive treatment. Intraperitoneal pioglitazone significantly reduced the nerve injury-induced up-regulation of cd11b, GFAP, and p-p38 in the dorsal horn, indicating a mechanism of action involving spinal microglia and/or astrocyte activation. Oral pioglitazone significantly reduced touch stimulus-evoked phospho-extracellular signal-related kinase (p-ERK) in lamina I-II, indicating a mechanism of action involving inhibition of central sensitization. We conclude that pioglitazone reduces spinal glial and stimulus-evoked p-ERK activation and that PPARγ activation blocks the development of and reduces established neuropathic pain.
Collapse
Affiliation(s)
- J Morgenweck
- Department of Physiology, University of Kentucky Research Foundation, Lexington, KY 40536, USA
| | | | | | | | | |
Collapse
|
38
|
Uher T, Bob P. Neuropathic Pain, Depressive Symptoms, and C-Reactive Protein in Sciatica Patients. Int J Neurosci 2012; 123:204-8. [DOI: 10.3109/00207454.2012.746335] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Hasegawa-Moriyama M, Ohnou T, Godai K, Kurimoto T, Nakama M, Kanmura Y. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuates postincisional pain by regulating macrophage polarization. Biochem Biophys Res Commun 2012; 426:76-82. [DOI: 10.1016/j.bbrc.2012.08.039] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/08/2012] [Indexed: 10/28/2022]
|
40
|
Iannitti T, Graham A, Dolan S. Increased central and peripheral inflammation and inflammatory hyperalgesia in Zucker rat model of leptin receptor deficiency and genetic obesity. Exp Physiol 2012; 97:1236-45. [PMID: 22523380 DOI: 10.1113/expphysiol.2011.064220] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This study investigated whether sensitivity to nociceptive stimuli is altered in obese rats using established models of inflammatory pain, and using real-time PCR, profiled alterations in expression of key adipokine and inflammatory mediator mRNA (adiponectin, tumor necrosis factor-α, interleukin-1β, cyclooxygenase-2, inducible nitric oxide synthase (iNOS)) in spinal cord with obesity. Responses to thermal and mechanical stimulation of the hindpaw and paw oedema were assessed in adult male Zucker fatty rats (fa/fa) and their lean littermates (fa/-; n = 6-9 per group) in the absence of inflammation (acute nociception), then in response to intradermal hindpaw injection of carrageenan (3%; 50 μl) or capsaicin (10 μg; 50 μl) or hindpaw incision. The analgesic potency of morphine (1, 2.5 or 5 mg kg(-1) or vehicle; s.c.) was also assessed. Acute nociception was unaltered in obese animals, but following carrageenan-induced inflammation the obese rats were significantly more sensitive to mechanical and thermal stimulation of the inflamed paw, and displayed greater paw oedema. No difference in the capsaicin- or paw-incision-induced pain sensitivity or in the analgesic potency of morphine was observed between groups. Levels of adiponectin and inducible nitric oxide synthase mRNA were downregulated in spinal cord from obese rats, whereas tumour necrosis factor-α mRNA was upregulated; interleukin-1β and cyclo-oxygenase were unchanged. The increased pain sensitivity and inflammatory response together with changes in spinal adipokine expression in obese rats fit well with the hypothesis that obesity is a chronic low-grade inflammatory disorder, producing a state where responses to subsequent inflammatory challenge are potentiated.
Collapse
Affiliation(s)
- Tommaso Iannitti
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | | | | |
Collapse
|
41
|
Graeber MB, Christie MJ. Multiple mechanisms of microglia: a gatekeeper's contribution to pain states. Exp Neurol 2012; 234:255-61. [PMID: 22273537 DOI: 10.1016/j.expneurol.2012.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 12/20/2011] [Accepted: 01/05/2012] [Indexed: 12/30/2022]
Abstract
Microglia are gatekeepers in the CNS for a wide range of pathological stimuli and they blow the whistle when things go wrong. Collectively, microglia form a CNS tissue alarm system (Kreutzberg's "sensor of pathology"), and their involvement in physiological pain is in line with this function. However, pathological neuropathic pain is characterized by microglial activation that is unwanted and considered to contribute to or even cause tactile allodynia, hyperalgesia and spontaneous pain. Such abnormal microglial behavior seems likely due to an as yet ill-understood disturbance of microglial functions unrelated to inflammation. The idea that microglia have roles in the CNS that differ from those of peripheral macrophages has gained momentum with the discovery of their separate, pre-hematopoietic lineage during embryonic development and their direct interactions with synapses.
Collapse
Affiliation(s)
- Manuel B Graeber
- Brain Tumor Research Laboratories, The Brain and Mind Research Institute, University of Sydney, Sydney, Australia.
| | | |
Collapse
|