1
|
Patilas C, Varsamos I, Galanis A, Vavourakis M, Zachariou D, Marougklianis V, Kolovos I, Tsalimas G, Karampinas P, Kaspiris A, Vlamis J, Pneumaticos S. The Role of Interleukin-10 in the Pathogenesis and Treatment of a Spinal Cord Injury. Diagnostics (Basel) 2024; 14:151. [PMID: 38248028 PMCID: PMC10814517 DOI: 10.3390/diagnostics14020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that often leads to severe and permanent neurological deficits. The complex pathophysiology of an SCI involves a cascade of events, including inflammation, oxidative stress, and secondary injury processes. Among the myriad of molecular players involved, interleukin-10 (IL-10) emerges as a key regulator with the potential to modulate both the inflammatory response and promote neuroprotection. This comprehensive review delves into the intricate interplay of IL-10 in the pathogenesis of an SCI and explores its therapeutic implications in the quest for effective treatments. IL-10 has been found to regulate inflammation, oxidative stress, neuronal apoptosis, and glial scars after an SCI. Its neuroprotective properties have been evaluated in a plethora of animal studies. IL-10 administration, either isolated or in combination with other molecules or biomaterials, has shown neuroprotective effects through a reduction in inflammation, the promotion of tissue repair and regeneration, the modulation of glial scar formation, and improved functional outcomes. In conclusion, IL-10 emerges as a pivotal player in the pathogenesis and treatment of SCIs. Its multifaceted role in modulating inflammation, oxidative stress, neuronal apoptosis, glial scars, and neuroprotection positions IL-10 as a promising therapeutic target. The ongoing research exploring various strategies for harnessing the potential of IL-10 offers hope for the development of effective treatments that could significantly improve outcomes for individuals suffering from spinal cord injuries. As our understanding of IL-10's intricacies deepens, it opens new avenues for innovative and targeted therapeutic interventions, bringing us closer to the goal of alleviating the profound impact of SCIs on patients' lives.
Collapse
Affiliation(s)
| | | | | | - Michail Vavourakis
- 3rd Department of Orthopaedic Surgery, National & Kapodistrian University of Athens, KAT General Hospital, 14561 Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Dugan EA, Schachner B, Jergova S, Sagen J. Intensive Locomotor Training Provides Sustained Alleviation of Chronic Spinal Cord Injury-Associated Neuropathic Pain: A Two-Year Pre-Clinical Study. J Neurotrauma 2021; 38:789-802. [PMID: 33218293 DOI: 10.1089/neu.2020.7378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuropathic pain often accompanies the functional deficits associated with spinal cord injury (SCI) and further reduces a patient's quality of life. Clinical and pre-clinical research is beginning to highlight the beneficial role that rehabilitative therapies such as locomotor training can have not only on functional recovery but also on chronic pain management. Our group has previously developed an intensive locomotor training (ILT) treadmill protocol on rats that reduced SCI neuropathic pain symptoms for at least 3 months. We have extended these findings in the current study to evaluate the ability of regular ILT regimen over a 2 year period post-SCI to maintain neuropathic pain reduction. To assess this, the rat clip compression SCI model (T7/8) was used and treadmill training was initiated starting 4 weeks after SCI and continuing through the duration of the study. Results showed continued suppression of SCI neuropathic pain responses (reduced mechanical, heat, and cold hypersensitivity throughout the entire time course of the study). In contrast, non-exercised rats showed consistent and sustained neuropathic pain responses during this period. In addition, prolonged survival and improved locomotor outcomes were observed in rats undergoing ILT as the study longevity progressed. Potential contributory mechanisms underlying beneficial effects of ILT include reduced inflammation and restoration of anti-nociceptive inhibitory processes as indicated by neurochemical assays in spinal tissue of remaining rats at 2 years post-SCI. The benefits of chronic ILT suggest that long-term physical exercise therapy can produce powerful and prolonged management of neuropathic pain, partly through sustained reduction of spinal pathological processes.
Collapse
Affiliation(s)
- Elizabeth A Dugan
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Benjamin Schachner
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Stanislava Jergova
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Jacqueline Sagen
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| |
Collapse
|
3
|
Andrés-Rodríguez L, Borràs X, Feliu-Soler A, Pérez-Aranda A, Rozadilla-Sacanell A, Arranz B, Montero-Marin J, García-Campayo J, Angarita-Osorio N, Maes M, Luciano JV. Machine Learning to Understand the Immune-Inflammatory Pathways in Fibromyalgia. Int J Mol Sci 2019; 20:ijms20174231. [PMID: 31470635 PMCID: PMC6747258 DOI: 10.3390/ijms20174231] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/24/2019] [Accepted: 08/26/2019] [Indexed: 12/30/2022] Open
Abstract
Fibromyalgia (FM) is a chronic syndrome characterized by widespread musculoskeletal pain, and physical and emotional symptoms. Although its pathophysiology is largely unknown, immune-inflammatory pathways may be involved. We examined serum interleukin (IL)-6, high sensitivity C-reactive protein (hs-CRP), CXCL-8, and IL-10 in 67 female FM patients and 35 healthy women while adjusting for age, body mass index (BMI), and comorbid disorders. We scored the Fibromyalgia Severity Score, Widespread Pain Index (WPI), Symptom Severity Scale (SSS), Hospital Anxiety (HADS-A), and Depression Scale and the Perceived Stress Scale (PSS-10). Clinical rating scales were significantly higher in FM patients than in controls. After adjusting for covariates, IL-6, IL-10, and CXCL-8 were lower in FM than in HC, whereas hs-CRP did not show any difference. Binary regression analyses showed that the diagnosis FM was associated with lowered IL-10, quality of sleep, aerobic activities, and increased HADS-A and comorbidities. Neural networks showed that WPI was best predicted by quality of sleep, PSS-10, HADS-A, and the cytokines, while SSS was best predicted by PSS-10, HADS-A, and IL-10. Lowered levels of cytokines are associated with FM independently from confounders. Lowered IL-6 and IL-10 signaling may play a role in the pathophysiology of FM.
Collapse
Affiliation(s)
- Laura Andrés-Rodríguez
- Group of Psychological Research in Fibromyalgia & Chronic Pain (AGORA), Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Teaching, Research & Innovation Unit, Parc Sanitari Sant Joan de Déu, 08830 St. Boi de Llobregat, Spain
- Primary Care Prevention and Health Promotion Research Network, RedIAPP, 28013 Madrid, Spain
- Faculty of Psychology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès Barcelona, Spain
| | - Xavier Borràs
- Group of Psychological Research in Fibromyalgia & Chronic Pain (AGORA), Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Faculty of Psychology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès Barcelona, Spain
| | - Albert Feliu-Soler
- Group of Psychological Research in Fibromyalgia & Chronic Pain (AGORA), Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain.
- Teaching, Research & Innovation Unit, Parc Sanitari Sant Joan de Déu, 08830 St. Boi de Llobregat, Spain.
- Primary Care Prevention and Health Promotion Research Network, RedIAPP, 28013 Madrid, Spain.
- Faculty of Psychology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès Barcelona, Spain.
| | - Adrián Pérez-Aranda
- Group of Psychological Research in Fibromyalgia & Chronic Pain (AGORA), Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain.
- Teaching, Research & Innovation Unit, Parc Sanitari Sant Joan de Déu, 08830 St. Boi de Llobregat, Spain.
- Primary Care Prevention and Health Promotion Research Network, RedIAPP, 28013 Madrid, Spain.
| | | | - Belén Arranz
- Parc Sanitari Sant Joan de Déu, CIBERSAM, 08950 Sant Boi de llobregat, Spain
| | - Jesús Montero-Marin
- Primary Care Prevention and Health Promotion Research Network, RedIAPP, 28013 Madrid, Spain
| | - Javier García-Campayo
- Primary Care Prevention and Health Promotion Research Network, RedIAPP, 28013 Madrid, Spain
- Department of Psychiatry, Miguel Servet Hospital, Aragon Institute of Health Sciences (I+CS), 50009 Zaragoza, Spain
| | - Natalia Angarita-Osorio
- Group of Psychological Research in Fibromyalgia & Chronic Pain (AGORA), Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
- Teaching, Research & Innovation Unit, Parc Sanitari Sant Joan de Déu, 08830 St. Boi de Llobregat, Spain
| | - Michael Maes
- Department of Psychiatry, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Juan V Luciano
- Group of Psychological Research in Fibromyalgia & Chronic Pain (AGORA), Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain.
- Teaching, Research & Innovation Unit, Parc Sanitari Sant Joan de Déu, 08830 St. Boi de Llobregat, Spain.
- Primary Care Prevention and Health Promotion Research Network, RedIAPP, 28013 Madrid, Spain.
| |
Collapse
|
4
|
Zhao J, Gao QY. TMEM16F inhibition limits pain-associated behavior and improves motor function by promoting microglia M2 polarization in mice. Biochem Biophys Res Commun 2019; 517:603-610. [PMID: 31409484 DOI: 10.1016/j.bbrc.2019.07.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) leads to sensorimotor deficits and autonomic changes. Macrophages and microglia could be polarized into the classically activated pro-inflammatory M1 phenotype or the alternatively activated anti-inflammatory M2 phenotype. Transmembrane protein with unknown function 16F (TMEM16F) exhibits functional diversity and may contribute to microglial function. However, the effects of TMEM16F on the modulation of macrophage/microglial polarization are still not fully understood. In the study, TMEM16F up-regulation was detected after SCI in mice, and TMEM16F protein was found in macrophages/microglia in injured spinal cord sections. Depletion of TMEM16F improved motor function in male mice with SCI. M1-type macrophages/microglia accumulated in lower numbers in the injured spinal cord of TMEM16F-knockout (KO) mice. M2 polarization inhibited by SCI was improved in mice with TMEM16F deficiency. TMEM16F deletion also attenuated microglial/macrophage pro-inflammatory response. Furthermore, significant down-regulation of A disintegrin and metalloprotease 17 (ADAM17) was observed in TMEM16F-KO mice. Importantly, TMEM16F-promoted M1 polarization and -inhibited M1 polarization were largely associated with the suppression of ADAM17. Overall, our findings provided new insights into the regulatory mechanisms of macrophage/microglial polarization, thereby possibly facilitating the development of new therapeutic strategies for SCI through the regulation of TMEM16F/ADAM17 signaling.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Orthopedics Spinal Ward, Xi'an Hospital of Traditional Chinese Medicine, Xi'an City, Shaanxi Province, 710021, China
| | - Quan-You Gao
- Department of Orthopaedics, Tangdu Hospital Second Affiliated Hospital of Air Force Military Medical University, Xi'an City, Shaanxi Province, 710038, China.
| |
Collapse
|
5
|
Sabirzhanov B, Li Y, Coll-Miro M, Matyas JJ, He J, Kumar A, Ward N, Yu J, Faden AI, Wu J. Inhibition of NOX2 signaling limits pain-related behavior and improves motor function in male mice after spinal cord injury: Participation of IL-10/miR-155 pathways. Brain Behav Immun 2019; 80:73-87. [PMID: 30807841 PMCID: PMC6660361 DOI: 10.1016/j.bbi.2019.02.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/11/2019] [Accepted: 02/22/2019] [Indexed: 12/19/2022] Open
Abstract
NADPH oxidase (NOX2) is an enzyme that induces reactive oxygen species (ROS) and serves as a switch between the pro-inflammatory and neurorestorative microglial/macrophage phenotypes; such changes play an important role in neuropathic pain and motor dysfunction. Increased NOX2 expression after spinal cord injury (SCI) has been reported, and inhibition of NOX2 improves motor function. However, the underlying mechanisms of NOX2 in post-traumatic pain and motor deficit remain unexplored. In the present study, we report that depletion of NOX2 (NOX2-/-) or inhibition of NOX2 using NOX2ds-tat significantly reduced mechanical/thermal cutaneous hypersensitivity and motor dysfunction after moderate contusion SCI at T10 in male mice. Western blot (WB, 3 mm lesion area) and immunohistochemistry (IHC) showed that SCI elevates NOX2 expression predominantly in microglia/macrophages up to 8 weeks post-injury. Deletion of NOX2 significantly reduced CD11b+/CD45hiF4/80+ macrophage infiltration at 24 h post-injury detected by flow cytometry and 8-OHG+ ROS production at 8 weeks post-injury by IHC in both lesion area and lumbar enlargement. NOX2 deficiency also altered microglial/macrophage pro-inflammatory and anti-inflammatory balance towards the neurorestorative response. WB analysis showed robust increase of Arginase-1 and YM1 proteins in NOX2-/- mice. Furthermore, qPCR analysis showed significant up-regulation of anti-inflammatory cytokine IL-10 levels in NOX2-/- mice, associated with reduced microRNA-155 expression. These findings were confirmed in CD11b+ microglia/macrophages isolated from spinal cord at 3 days post-injury. Taken together, our data suggest an important role for IL-10/miR-155 pathway in regulating NOX2-mediated SCI-dysfunction. Thus, specific targeting of NOX2 may provide an effective strategy for treating neurological dysfunction in SCI patients.
Collapse
Affiliation(s)
- Boris Sabirzhanov
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Marino Coll-Miro
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Jessica J. Matyas
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Alok Kumar
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Nicole Ward
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Jingwen Yu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201 USA,University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201 USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201 USA.
| |
Collapse
|
6
|
Chio JCT, Wang J, Badner A, Hong J, Surendran V, Fehlings MG. The effects of human immunoglobulin G on enhancing tissue protection and neurobehavioral recovery after traumatic cervical spinal cord injury are mediated through the neurovascular unit. J Neuroinflammation 2019; 16:141. [PMID: 31288834 PMCID: PMC6615094 DOI: 10.1186/s12974-019-1518-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/11/2019] [Indexed: 12/30/2022] Open
Abstract
Background Spinal cord injury (SCI) is a condition with few effective treatment options. The blood-spinal cord barrier consists of pericytes, astrocytes, and endothelial cells, which are collectively termed the neurovascular unit. These cells support spinal cord homeostasis by expressing tight junction proteins. Physical trauma to the spinal cord disrupts the barrier, which leads to neuroinflammation by facilitating immune cell migration to the damaged site in a process involving immune cell adhesion. Immunosuppressive strategies, including methylprednisolone (MPSS), have been investigated to treat SCI. However, despite some success, MPSS has the potential to increase a patient’s susceptibility to wound infection and impaired wound healing. Hence, immunomodulation may be a more attractive approach than immunosuppression. Approved for modulating neuroinflammation in certain disorders, including Guillain-Barre syndrome, intravenous administration of human immunoglobulin G (hIgG) has shown promise in the setting of experimental SCI, though the optimal dose and mechanism of action remain undetermined. Methods Female adult Wistar rats were subjected to moderate-severe clip compression injury (35 g) at the C7-T1 level and randomized to receive a single intravenous (IV) bolus of hIgG (0.02, 0.2, 0.4, 1, 2 g/kg), MPSS (0.03 g/kg), or control buffer at 15 min post-SCI. At 24 h and 6 weeks post-SCI, molecular, histological, and neurobehavioral effects of hIgG were analyzed. Results At 24 h post-injury, human immunoglobulin G co-localized with spinal cord pericytes, astrocytes, and vessels. hIgG (2 g/kg) protected the spinal cord neurovasculature after SCI by increasing tight junction protein expression and reducing inflammatory enzyme expression. Improvements in vascular integrity were associated with changes in spinal cord inflammation. Interestingly, hIgG (2 g/kg) increased serum expression of inflammatory cytokines and co-localized (without decreasing protein expression) with spinal cord vascular cell adhesion molecule-1, a protein used by immune cells to enter into inflamed tissue. Acute molecular benefits of hIgG (2 g/kg) led to greater tissue preservation, functional blood flow, and neurobehavioral recovery at 6 weeks post-SCI. Importantly, the effects of hIgG (2 g/kg) were superior to control buffer and hIgG (0.4 g/kg), and comparable with MPSS (0.03 g/kg). Conclusions hIgG (2 g/kg) is a promising therapeutic approach to mitigate secondary pathology in SCI through antagonizing immune cell infiltration at the level of the neurovascular unit.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jian Wang
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada
| | - Anna Badner
- Sue and Bill Gross Stem Cell Research Centre, University of California, 845 Health Sciences Road, Irvine, CA, 92617, USA
| | - James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | | | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada. .,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada. .,University of Toronto, Toronto, Ontario, Canada. .,Gerry and Tootsie Halbert Chair in Neural Repair and Regeneration, University of Toronto, Toronto, Canada. .,Krembil Neuroscience Program, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, Ontario, M5T 2S8, Canada.
| |
Collapse
|
7
|
Badner A, Vidal PM, Hong J, Hacker J, Fehlings MG. Endogenous Interleukin-10 Deficiency Exacerbates Vascular Pathology in Traumatic Cervical Spinal Cord Injury. J Neurotrauma 2019; 36:2298-2307. [PMID: 30843463 DOI: 10.1089/neu.2018.6081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although the majority of traumatic spinal cord injuries (SCIs) take place at the cervical level, pre-clinical studies have been disproportionally focused on thoracic insults. With differences in anatomy, physiology, and immune response between spinal cord levels, there is evidence that injury pathophysiology may vary, requiring tailored treatment paradigms. Further, as only a few therapies have been successfully translated to the clinic, cervical models are increasingly recognized as essential for the characterization of trauma and therapy. Using a novel and clinically relevant cervical contusion-compression mouse model of bilateral incomplete injury, this study aimed to assess the role of interleukin10 (IL-10), a potent cytokine with broad anti-inflammatory effects, in SCI vascular pathology. While the effects of IL-10 loss have been previously evaluated, the vascular changes are poorly characterized. Here, using in vivo high-resolution ultrasound imaging, we demonstrate that IL-10 deficiency is associated with increased acute vascular damage. Importantly, the loss of endogenous IL-10 led to significant differences in the acute systemic response to SCI, specifically the circulating levels of IL-12 (p70), LIX (CXCL5), IL-1β, tumor necrosis factor (TNF)-α, and IL-6 relative to genotype sham controls. These effects also fostered modest impairments in long-term functional recovery, assessed by the Basso Mouse Scale, as well as histological outcomes.
Collapse
Affiliation(s)
- Anna Badner
- 1Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,2Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Pia M Vidal
- 1Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - James Hong
- 1Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,2Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Justin Hacker
- 1Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael G Fehlings
- 1Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,2Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Jurga AM, Rojewska E, Makuch W, Mika J. Lipopolysaccharide from Rhodobacter sphaeroides (TLR4 antagonist) attenuates hypersensitivity and modulates nociceptive factors. PHARMACEUTICAL BIOLOGY 2018; 56:275-286. [PMID: 29656686 PMCID: PMC6130482 DOI: 10.1080/13880209.2018.1457061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Accumulating evidence has demonstrated that Toll-like receptors (TLRs), especially TLR4 localized on microglia/macrophages, may play a significant role in nociception. OBJECTIVE We examine the role of TLR4 in a neuropathic pain model. Using behavioural/biochemical methods, we examined the influence of TLR4 antagonist on levels of hypersensitivity and nociceptive factors whose contribution to neuropathy development has been confirmed. MATERIALS AND METHODS Behavioural (von Frey's/cold plate) tests were performed with Wistar male rats after intrathecal administration of a TLR4 antagonist (LPS-RS ULTRAPURE (LPS-RSU), 20 μG: lipopolysaccharide from Rhodobacter sphaeroides, InvivoGen, San Diego, CA) 16 H and 1 h before chronic constriction injury (cci) to the sciatic nerve and then daily for 7 d. three groups were used: an intact group and two cci-exposed groups that received vehicle or LPS-RSU. tissue [spinal cord/dorsal root ganglia (DRG)] for western blot analysis was collected on day 7. RESULTS The pharmacological blockade of TLR4 diminished mechanical (from ca. 40% to 16% that in the INTACT group) and thermal (from ca. 51% to 32% that in the INTACT group) hypersensitivity despite the enhanced activation of IBA-1-positive cells in DRG. Moreover, LPS-RSU changed the ratio between IL-18/IL-18BP and MMP-9/TIMP-1 in favour of the increase of antinociceptive factors IL-18BP (25%-spinal; 96%-DRG) and TIMP-1 (15%-spinal; 50%-DRG) and additionally led to an increased IL-6 (40%-spinal; 161%-DRG), which is known to have analgesic properties in neuropathy. CONCLUSIONS Our results provide evidence that LPS-RSU influences pain through the expression of TLR4. TLR4 blockade has analgesic properties and restores the balance between nociceptive factors, which indicates its engagement in neuropathy development.
Collapse
Affiliation(s)
- Agnieszka M. Jurga
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
- CONTACT Joanna MikaDepartment of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Str., 31343Krakow, Poland
| |
Collapse
|
9
|
Ren H, Chen X, Tian M, Zhou J, Ouyang H, Zhang Z. Regulation of Inflammatory Cytokines for Spinal Cord Injury Repair Through Local Delivery of Therapeutic Agents. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800529. [PMID: 30479916 PMCID: PMC6247077 DOI: 10.1002/advs.201800529] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/06/2018] [Indexed: 05/29/2023]
Abstract
The balance of inflammation is critical to the repair of spinal cord injury (SCI), which is one of the most devastating traumas in human beings. Inflammatory cytokines, the direct mediators of local inflammation, have differential influences on the repair of the injured spinal cord. Some inflammatory cytokines are demonstrated beneficial to spinal cord repair in SCI models, while some detrimental. Various animal researches have revealed that local delivery of therapeutic agents efficiently regulates inflammatory cytokines and promotes repair from SCI. Quite a few clinical studies have also shown the promotion of repair from SCI through regulation of inflammatory cytokines. However, local delivery of a single agent affects only a part of the inflammatory cytokines that need to be regulated. Meanwhile, different individuals have differential profiles of inflammatory cytokines. Therefore, future studies may aim to develop personalized strategies of locally delivered therapeutic agent cocktails for effective and precise regulation of inflammation, and substantial functional recovery from SCI.
Collapse
Affiliation(s)
- Hao Ren
- The Third Affiliated Hospital of Guangzhou Medical UniversityNo. 63 Duobao RoadGuangzhou510150P. R. China
| | - Xuri Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Mengya Tian
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Jing Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineSchool of Basic Medical ScienceZhejiang UniversityNo. 866 Yuhangtang RoadHangzhou310058P. R. China
| | - Zhiyong Zhang
- Translational Research Center for Regenerative Medicine and 3D Printing TechnologiesGuangzhou Medical UniversityNo. 63 Duobao RoadGuangzhou510150P. R. China
| |
Collapse
|
10
|
Saxena N, Won J, Choi S, Singh AK, Singh I. S-nitrosoglutathione reductase (GSNOR) inhibitor as an immune modulator in experimental autoimmune encephalomyelitis. Free Radic Biol Med 2018; 121:57-68. [PMID: 29694854 PMCID: PMC6083447 DOI: 10.1016/j.freeradbiomed.2018.04.558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Abstract
We previously reported that S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier, attenuated TH17-mediated immune responses in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Cellular GSNO homeostasis is regulated via its synthesis by reaction between nitric oxide and glutathione and its enzymatic catabolism by GSNO reductase (GSNOR). In this study, we evaluated potential of reversible inhibitor of GSNOR (N6022) in comparison with exogenous GSNO in immunopathogenesis of EAE. Daily treatment of EAE mice with N6022 or exogenous GSNO significantly attenuated the clinical disease of EAE, but N6022 treatment showed greater efficacy than GSNO. Both N6022 and exogenous GSNO treatments increased the spleen levels of GSNO, as documented by increased protein-associated S-nitrosothiols, and inhibited polarization and CNS effector function of proinflammatory TH17 cells while inducing the polarization and CNS effector function of anti-inflammatory CD4+ CD25+ FOXP3- regulatory T (Treg) cells. Moreover, N6022 further attenuated TH1 while inducing TH2 and CD4+ CD25+ FOXP3+ Treg in their polarization and CNS effector functions. Similar to GSNO, the N6022 treatment protected against the EAE disease induced demyelination. However, neither exogenous GSNO nor N6022 treatment did not cause significant systemic lymphopenic effect as compared to FTY720. Taken together, these data document that optimization of cellular GSNO homeostasis by GSNOR inhibitor (N6022) in NO metabolizing cells attenuates EAE disease via selective inhibition of pro-inflammatory subsets of CD4+ cells (TH1/TH17) while upregulating anti-inflammatory subsets of CD4+ cells (TH2/Treg) without causing lymphopenic effects and thus offers a potential treatment option for MS/EAE.
Collapse
MESH Headings
- Alcohol Dehydrogenase/antagonists & inhibitors
- Animals
- Benzamides/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Mice
- Mice, Inbred C57BL
- Protein S/metabolism
- Pyrroles/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/drug effects
- Th1 Cells/enzymology
- Th1 Cells/immunology
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Seungho Choi
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| |
Collapse
|
11
|
García E, Silva-García R, Flores-Romero A, Blancas-Espinoza L, Rodríguez-Barrera R, Ibarra A. The Severity of Spinal Cord Injury Determines the Inflammatory Gene Expression Pattern after Immunization with Neural-Derived Peptides. J Mol Neurosci 2018; 65:190-195. [PMID: 29796836 DOI: 10.1007/s12031-018-1077-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/27/2018] [Indexed: 11/28/2022]
Abstract
Previous studies revealed that the intensity of spinal cord injury (SCI) plays a key role in the therapeutic effects induced by immunizing with neural-derived peptides (INDP), as severe injuries abolish the beneficial effects induced by INDP. In the present study, we analyzed the expression of some inflammation-related genes (IL6, IL12, IL-1β, IFNɣ, TNFα, IL-10, IL-4, and IGF-1) by quantitative PCR in rats subjected to SCI and INDP. We investigated the expression of these genes after a moderate or severe contusion. In addition, we evaluated the effect of INDP by utilizing two different peptides: A91 and Cop-1. After moderate injury, both A91 and Cop-1 elicited a pattern of genes characterized by a significant reduction of IL6, IL1β, and TNFα but an increase in IL10, IL4, and IGF-1 expression. There was no effect on IL-12 and INFɣ. In contrast, the opposite pattern was observed when rats were subjected to a severe spinal cord contusion. Immunization with either peptide caused a significant increase in the expression of IL-12, IL-1β, IFNɣ (pro-inflammatory genes), and IGF-1. There was no effect on IL-4 and IL-10 compared to controls. After a moderate SCI, INDP reduced pro-inflammatory gene expression and generated a microenvironment prone to neuroprotection. Nevertheless, severe injury elicits the expression of pro-inflammatory genes that could be aggravated by INDP. These findings correlate with our previous results demonstrating that severe injury inhibits the beneficial effects of protective autoimmunity.
Collapse
Affiliation(s)
- Elisa García
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, C.P. 52786, Huixquilucan Edo. de México, México.,Centro de Investigación del Proyecto CAMINA A.C., 14050, Mexico City, Mexico
| | - Raúl Silva-García
- Departamento de Inmunología, CMN Siglo XXI, 06720, Mexico City, Mexico
| | - Adrian Flores-Romero
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, C.P. 52786, Huixquilucan Edo. de México, México.,Centro de Investigación del Proyecto CAMINA A.C., 14050, Mexico City, Mexico
| | | | - Roxana Rodríguez-Barrera
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, C.P. 52786, Huixquilucan Edo. de México, México.,Centro de Investigación del Proyecto CAMINA A.C., 14050, Mexico City, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, C.P. 52786, Huixquilucan Edo. de México, México. .,Centro de Investigación del Proyecto CAMINA A.C., 14050, Mexico City, Mexico.
| |
Collapse
|
12
|
Uhde AK, Ciurkiewicz M, Herder V, Khan MA, Hensel N, Claus P, Beckstette M, Teich R, Floess S, Baumgärtner W, Jung K, Huehn J, Beineke A. Intact interleukin-10 receptor signaling protects from hippocampal damage elicited by experimental neurotropic virus infection of SJL mice. Sci Rep 2018; 8:6106. [PMID: 29666403 PMCID: PMC5904160 DOI: 10.1038/s41598-018-24378-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/27/2018] [Indexed: 12/24/2022] Open
Abstract
Theiler’s murine encephalomyelitis virus (TMEV) infection represents an experimental mouse model to study hippocampal damage induced by neurotropic viruses. IL-10 is a pleiotropic cytokine with profound anti-inflammatory properties, which critically controls immune homeostasis. In order to analyze IL-10R signaling following virus-induced polioencephalitis, SJL mice were intracerebrally infected with TMEV. RNA-based next generation sequencing revealed an up-regulation of Il10, Il10rα and further genes involved in IL-10 downstream signaling, including Jak1, Socs3 and Stat3 in the brain upon infection. Subsequent antibody-mediated blockade of IL-10R signaling led to enhanced hippocampal damage with neuronal loss and increased recruitment of CD3+ T cells, CD45R+ B cells and an up-regulation of Il1α mRNA. Increased expression of Tgfβ and Foxp3 as well as accumulation of Foxp3+ regulatory T cells and arginase-1+ macrophages/microglia was detected in the hippocampus, representing a potential compensatory mechanism following disturbed IL-10R signaling. Additionally, an increased peripheral Chi3l3 expression was found in spleens of infected mice, which may embody reactive regulatory mechanisms for prevention of excessive immunopathology. The present study highlights the importance of IL-10R signaling for immune regulation and its neuroprotective properties in the context of an acute neurotropic virus infection.
Collapse
Affiliation(s)
- Ann-Kathrin Uhde
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Muhammad Akram Khan
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany.,Department of Pathobiology, Faculty of Veterinary & Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Peter Claus
- Center for Systems Neuroscience, Hannover, Germany.,Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Michael Beckstette
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - René Teich
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany.,Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
13
|
Sustained reversal of central neuropathic pain induced by a single intrathecal injection of adenosine A 2A receptor agonists. Brain Behav Immun 2018; 69:470-479. [PMID: 29366930 DOI: 10.1016/j.bbi.2018.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/19/2023] Open
Abstract
Central neuropathic pain is a debilitating outcome of spinal cord injury (SCI) and current treatments to alleviate this pain condition are ineffective. A growing body of literature suggests that activating adenosine A2A receptors (A2ARs) decreases the production of proinflammatory cytokines and increases the production of anti-inflammatory cytokines. Here, the effect of administering intrathecal A2AR agonists on central neuropathic pain was measured using hindpaw mechanical allodynia in a rat model of SCI termed spinal neuropathic avulsion pain (SNAP). Other models of SCI cause extensive damage to the spinal cord, resulting in paralysis and health problems. SNAP rats with unilateral low thoracic (T13)/high lumbar (L1) dorsal root avulsion develop below-level bilateral allodynia, without concomitant motor or health problems. A single intrathecal injection of the A2AR agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine HCl (CGS21680) reversed SCI-induced allodynia for at least 6 weeks. The reversal is likely in part mediated by interleukin (IL)-10, as intrathecally administering neutralizing IL-10 antibodies 1 week after CGS21680 abolished the anti-allodynic effect of CGS21680. Dorsal spinal cord tissue from the ipsilateral site of SCI (T13/L1) was assayed 1 and 6 weeks after CGS21680 for IL-10, CD11b, and tumor necrosis factor (TNF) gene expression. CGS21680 treatment did not change IL-10 gene expression but did significantly decrease CD11b and TNF gene expression at both timepoints. A second A2AR agonist, 4-(3-(6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)prop-2-ynyl)piperidine-1-carboxylic acid methyl ester (ATL313), was also able to significantly prevent and reverse SCI-induced allodynia for several weeks after a single intrathecal injection, providing converging lines of evidence of A2AR involvement. The enduring pain reversal after a single intrathecal injection of A2AR agonists suggests that A2AR agonists could be exciting new candidates for treating SCI-induced central neuropathic pain.
Collapse
|
14
|
Walters ET. How is chronic pain related to sympathetic dysfunction and autonomic dysreflexia following spinal cord injury? Auton Neurosci 2017; 209:79-89. [PMID: 28161248 DOI: 10.1016/j.autneu.2017.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/29/2022]
Abstract
Autonomic dysreflexia (AD) and neuropathic pain occur after severe injury to higher levels of the spinal cord. Mechanisms underlying these problems have rarely been integrated in proposed models of spinal cord injury (SCI). Several parallels suggest significant overlap of these mechanisms, although the relationships between sympathetic function (dysregulated in AD) and nociceptive function (dysregulated in neuropathic pain) are complex. One general mechanism likely to be shared is central sensitization - enhanced responsiveness and synaptic reorganization of spinal circuits that mediate sympathetic reflexes or that process and relay pain-related information to the brain. Another is enhanced sensory input to spinal circuits caused by extensive alterations in primary sensory neurons. Both AD and SCI-induced neuropathic pain are associated with spinal sprouting of peptidergic nociceptors that might increase synaptic input to the circuits involved in AD and SCI pain. In addition, numerous nociceptors become hyperexcitable, hypersensitive to chemicals associated with injury and inflammation, and spontaneously active, greatly amplifying sensory input to sensitized spinal circuits. As discussed with the aid of a preliminary functional model, these effects are likely to have mutually reinforcing relationships with each other, and with consequences of SCI-induced interruption of descending excitatory and inhibitory influences on spinal circuits, with SCI-induced inflammation in the spinal cord and in DRGs, and with activity in sympathetic fibers within DRGs that promotes local inflammation and spontaneous activity in sensory neurons. This model suggests that interventions selectively targeting hyperactivity in C-nociceptors might be useful for treating chronic pain and AD after high SCI.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
15
|
Genetic Ablation of Soluble TNF Does Not Affect Lesion Size and Functional Recovery after Moderate Spinal Cord Injury in Mice. Mediators Inflamm 2016; 2016:2684098. [PMID: 28070141 PMCID: PMC5192339 DOI: 10.1155/2016/2684098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/24/2016] [Accepted: 11/03/2016] [Indexed: 11/23/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is followed by an instant increase in expression of the microglial-derived proinflammatory cytokine tumor necrosis factor (TNF) within the lesioned cord. TNF exists both as membrane-anchored TNF (mTNF) and as cleaved soluble TNF (solTNF). We previously demonstrated that epidural administration of a dominant-negative inhibitor of solTNF, XPro1595, to the contused spinal cord resulted in changes in Iba1 protein expression in microglia/macrophages, decreased lesion volume, and improved locomotor function. Here, we extend our studies using mice expressing mTNF, but no solTNF (mTNFΔ/Δ), to study the effect of genetic ablation of solTNF on SCI. We demonstrate that TNF levels were significantly decreased within the lesioned spinal cord 3 days after SCI in mTNFΔ/Δ mice compared to littermates. This decrease did, however, not translate into significant changes in other pro- and anti-inflammatory cytokines (IL-10, IL-1β, IL-6, IL-5, IL-2, CXCL1, CCL2, or CCL5), despite a tendency towards increased IL-10 and decreased IL-1β, TNFR1, and TNFR2 levels in mTNFΔ/Δ mice. In addition, microglial and leukocyte infiltration, activation state (Iba1, CD11b, CD11c, CD45, and MHCII), lesion size, and functional outcome after moderate SCI were comparable between genotypes. Collectively, our data demonstrate that genetic ablation of solTNF does not significantly modulate postlesion outcome after SCI.
Collapse
|
16
|
Sordillo PP, Sordillo LA, Helson L. Bifunctional role of pro-inflammatory cytokines after traumatic brain injury. Brain Inj 2016; 30:1043-53. [DOI: 10.3109/02699052.2016.1163618] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Liao MF, Yeh SR, Lo AL, Chao PK, Lee YL, Hung YH, Lu KT, Ro LS. An early granulocyte colony-stimulating factor treatment attenuates neuropathic pain through activation of mu opioid receptors on the injured nerve. Sci Rep 2016; 6:25490. [PMID: 27180600 PMCID: PMC4867617 DOI: 10.1038/srep25490] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 04/18/2016] [Indexed: 12/31/2022] Open
Abstract
Several studies have shown that the mu opioid receptor (MOR) located in the peripheral nerves can be activated after nerve injury and that it attenuates peripheral nociceptive signals to the spinal dorsal horn. Various cytokines and phosphorylated-p38 (p-p38) activation in the dorsal horn also play an important role in neuropathic pain development. Granulocyte-colony stimulating factor (GCSF) is a growth factor that can stimulate granulocyte formation and has been shown to exert an analgesic effect on neuropathic pain through recruiting opioid-containing leukocytes to the injured nerve. However, the underlying mechanisms are not well understood. Herein, the results of behavior tests in addition to MOR levels in the injured sciatic nerve and the levels of p-p38 and various cytokines in the spinal dorsal horn were studied in vehicle-treated or GCSF-treated chronic constriction injured (CCI) rats at different time points (i.e., 1, 3, and 7 days, respectively) after nerve injury. The results showed that a single early systemic GCSF treatment after nerve injury can up-regulate MORs in the injured nerve, which can decrease peripheral nociceptive signals. Thereafter, those changes suppress the pro-inflammatory cytokine IL-6 but enhance the anti-inflammatory cytokine IL-4, followed by decreases in p-p38 in the dorsal horn, and thus further attenuate neuropathic pain.
Collapse
Affiliation(s)
- Ming-Feng Liao
- Department of Life Science, National Taiwan Normal University, 88, Ting-chou Rd., Sec. 4, Taipei, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 199, Tung Hwa North Rd., Taipei, Taiwan
| | - Shin-Rung Yeh
- College of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ai-Lun Lo
- Department of Neurology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 199, Tung Hwa North Rd., Taipei, Taiwan
| | - Po-Kuan Chao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yun-Lin Lee
- Department of Neurology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 199, Tung Hwa North Rd., Taipei, Taiwan
| | - Yu-Hui Hung
- Department of Neurology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 199, Tung Hwa North Rd., Taipei, Taiwan
| | - Kwok-Tung Lu
- Department of Life Science, National Taiwan Normal University, 88, Ting-chou Rd., Sec. 4, Taipei, Taiwan
| | - Long-Sun Ro
- Department of Neurology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, 199, Tung Hwa North Rd., Taipei, Taiwan
| |
Collapse
|
18
|
Thakur V, Gonzalez M, Pennington K, Chattopadhyay M. Viral vector mediated continuous expression of interleukin-10 in DRG alleviates pain in type 1 diabetic animals. Mol Cell Neurosci 2016; 72:46-53. [PMID: 26802537 DOI: 10.1016/j.mcn.2016.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/03/2015] [Accepted: 01/15/2016] [Indexed: 12/24/2022] Open
Abstract
Painful diabetic neuropathy is a common and difficult to treat complication of diabetes. A growing body of evidence implicates the role of inflammatory mediators in the damage to the peripheral axons and in the pathogenesis of neuropathic pain. Increased expression of pro-inflammatory cytokines such as interleukin (IL)-1β and tumor necrosis factor (TNF)-α in the peripheral nervous system suggests the possibility of change in pain perception in diabetes. In this study we investigated that continuous delivery of IL10 in the nerve fibers achieved by HSV vector mediated transduction of dorsal root ganglion (DRG) in animals with Type 1 diabetes, blocks the nociceptive and stress responses in the DRG neurons by reducing IL1β expression along with inhibition of phosphorylation of p38 MAPK (mitogen-activated protein kinase) and protein kinase C (PKC). The continuous expression of IL10 also alters Toll like receptor (TLR)-4 expression in the DRG with increased expression of heat shock protein (HSP)-70 in conjunction with the reduction of pain. Taken together, this study suggests that macrophage activation in the peripheral nervous system may be involved in the pathogenesis of pain in Type 1 diabetes and therapeutic benefits of HSV mediated local expression of IL10 in the DRG with the reduction of a number of proinflammatory cytokines, subsequently inhibits the development of painful neuropathy along with a decrease in stress associated markers in the DRG. This basic and preclinical study provides an important evidence for a novel treatment strategy that could lead to a clinical trial for what is currently a treatment resistant complication of diabetes.
Collapse
Affiliation(s)
- Vikram Thakur
- Center of Excellence in Diabetes and Obesity, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Mayra Gonzalez
- Center of Excellence in Diabetes and Obesity, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Kristen Pennington
- Center of Excellence in Diabetes and Obesity, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Munmun Chattopadhyay
- Center of Excellence in Diabetes and Obesity, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States.
| |
Collapse
|
19
|
Leung A, Gregory NS, Allen LAH, Sluka KA. Regular physical activity prevents chronic pain by altering resident muscle macrophage phenotype and increasing interleukin-10 in mice. Pain 2016; 157:70-79. [PMID: 26230740 PMCID: PMC4685958 DOI: 10.1097/j.pain.0000000000000312] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Regular physical activity in healthy individuals prevents development of chronic musculoskeletal pain; however, the mechanisms underlying this exercise-induced analgesia are not well understood. Interleukin-10 (IL-10), an antiinflammatory cytokine that can reduce nociceptor sensitization, increases during regular physical activity. Since macrophages play a major role in cytokine production and are present in muscle tissue, we propose that physical activity alters macrophage phenotype to increase IL-10 and prevent chronic pain. Physical activity was induced by allowing C57BL/6J mice free access to running wheels for 8 weeks and compared to sedentary mice with no running wheels. Using immunohistochemical staining of the gastrocnemius muscle to label regulatory (M2, secretes antiinflammatory cytokines) and classical (M1, secretes proinflammatory cytokines) macrophages, the percentage of M2-macrophages increased significantly in physically active mice (68.5% ± 4.6% of total) compared with sedentary mice (45.8% ± 7.1% of total). Repeated acid injections into the muscle enhanced mechanical sensitivity of the muscle and paw in sedentary animals, which does not occur in physically active mice; no sex differences occur in either sedentary or physically active mice. Blockade of IL-10 systemically or locally prevented the analgesia in physically active mice, ie, mice developed hyperalgesia. Conversely, sedentary mice pretreated systemically or locally with IL-10 had reduced hyperalgesia after repeated acid injections. Thus, these results suggest that regular physical activity increases the percentage of regulatory macrophages in muscle and that IL-10 is an essential mediator in the analgesia produced by regular physical activity.
Collapse
Affiliation(s)
- Audrey Leung
- University of Iowa Carver College of Medicine, Iowa City, IA, USA Neuroscience Graduate Program, University of Iowa, Iowa City, IA, USA Departments of Internal Medicine and Physical Therapy and Rehabilitation Science, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | | | | |
Collapse
|
20
|
Kabu S, Gao Y, Kwon BK, Labhasetwar V. Drug delivery, cell-based therapies, and tissue engineering approaches for spinal cord injury. J Control Release 2015; 219:141-154. [PMID: 26343846 DOI: 10.1016/j.jconrel.2015.08.060] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/23/2015] [Accepted: 08/31/2015] [Indexed: 12/28/2022]
Abstract
Spinal cord injury (SCI) results in devastating neurological and pathological consequences, causing major dysfunction to the motor, sensory, and autonomic systems. The primary traumatic injury to the spinal cord triggers a cascade of acute and chronic degenerative events, leading to further secondary injury. Many therapeutic strategies have been developed to potentially intervene in these progressive neurodegenerative events and minimize secondary damage to the spinal cord. Additionally, significant efforts have been directed toward regenerative therapies that may facilitate neuronal repair and establish connectivity across the injury site. Despite the promise that these approaches have shown in preclinical animal models of SCI, challenges with respect to successful clinical translation still remain. The factors that could have contributed to failure include important biologic and physiologic differences between the preclinical models and the human condition, study designs that do not mirror clinical reality, discrepancies in dosing and the timing of therapeutic interventions, and dose-limiting toxicity. With a better understanding of the pathobiology of events following acute SCI, developing integrated approaches aimed at preventing secondary damage and also facilitating neuroregenerative recovery is possible and hopefully will lead to effective treatments for this devastating injury. The focus of this review is to highlight the progress that has been made in drug therapies and delivery systems, and also cell-based and tissue engineering approaches for SCI.
Collapse
Affiliation(s)
- Shushi Kabu
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yue Gao
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Brian K Kwon
- Department of Orthopaedics, International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada V5Z 1M9
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
21
|
Song M, Jue SS, Cho YA, Kim EC. Comparison of the effects of human dental pulp stem cells and human bone marrow-derived mesenchymal stem cells on ischemic human astrocytes in vitro. J Neurosci Res 2015; 93:973-83. [PMID: 25663284 DOI: 10.1002/jnr.23569] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/27/2014] [Accepted: 01/12/2015] [Indexed: 12/11/2022]
Abstract
This study assesses the cytoprotective effects of human dental pulp stem cells (hDPSCs) and conditioned medium from hDPSCs (CM-hDPSCs) on ischemic human astrocytes (hAs) in vitro compared with human bone marrow-derived mesenchymal stem cells (hMSCs). Ischemia of hAs was induced by oxygen-glucose deprivation (OGD). CM-hDPSCs and hMSCs were collected after 48 hr of culture. Cell death was determined by 3-[4,5-dimethylthialzol-2-yl]-2,5-diphenyltetrazolium bromide and cellular ATP assays. The expression of glial fibrillary acidic protein (GFAP) and musashi-1 as markers of reactive astrogliosis was examined with immunochemical staining. mRNA expression and reactive oxygen species (ROS) were analyzed by RT-PCR and flow cytometry, respectively. OGD increased cytotoxicity in a time-dependent manner and decreased cellular ATP content concomitantly in hAs. Pretreatment and posttreatment with hDPSCs were associated with greater recovery from OGD-induced cytotoxicity in hAs compared with hMSCs. Similarly, CM-hDPSCs had a greater effect on OGD-induced cytotoxicity in a dose-dependent manner. Pre- and posttreatment with CM-hDPSCs or CM-hMSCs attenuated OGD-induced GFAP, nestin, and musashi-1 expression in hAs. Furthermore, treatment of cells with CM-hDPSCs and hMSCs blocked OGD-induced ROS production and interleukin-1ß upregulation. This study demonstrates for the first time that hDPSCs and CM-hDPSCs confer superior cytoprotection against cell death in an in vitro OGD model compared with hMSCs as shown by cell viability assay. Reactive gliosis, ROS production, and inflammatory mediators might contribute to this protective effect. Therefore, hDPSCs could represent an alternative source of cell therapy for ischemic stroke.
Collapse
Affiliation(s)
- Miyeoun Song
- Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
22
|
Li HD, Zhang QX, Mao Z, Xu XJ, Li NY, Zhang H. Exogenous interleukin-10 attenuates hyperoxia-induced acute lung injury in mice. Exp Physiol 2015; 100:331-40. [PMID: 25480159 DOI: 10.1113/expphysiol.2014.083337] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/25/2014] [Indexed: 01/11/2023]
Affiliation(s)
- Huai-Dong Li
- Department of Respiratory Disease; the 88th Hospital of the Chinese PLA; Taian 271000 China
| | - Qing-Xiang Zhang
- Department of Orthopedics; the 148th Hospital of the Chinese PLA; Zibo 255300 China
| | - Zhi Mao
- Department of Critical Care Medicine; the Chinese PLA General Hospital; Beijing 100853 China
| | - Xing-Jie Xu
- Department of TCM; The Affiliated Hospital of Taishan Medical College; Taian 271000 China
| | - Nai-Yi Li
- Department of Medical Services; the 88th Hospital of the Chinese PLA; Taian 271000 China
| | - Hui Zhang
- Department of Cardiology; the 88th Hospital of the Chinese PLA; Taian 271000 China
| |
Collapse
|
23
|
Walters ET. Neuroinflammatory contributions to pain after SCI: roles for central glial mechanisms and nociceptor-mediated host defense. Exp Neurol 2014; 258:48-61. [PMID: 25017887 DOI: 10.1016/j.expneurol.2014.02.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/23/2014] [Accepted: 02/02/2014] [Indexed: 12/30/2022]
Abstract
Neuropathic pain after spinal cord injury (SCI) is common, often intractable, and can be severely debilitating. A number of mechanisms have been proposed for this pain, which are discussed briefly, along with methods for revealing SCI pain in animal models, such as the recently applied conditioned place preference test. During the last decade, studies of animal models have shown that both central neuroinflammation and behavioral hypersensitivity (indirect reflex measures of pain) persist chronically after SCI. Interventions that reduce neuroinflammation have been found to ameliorate pain-related behavior, such as treatment with agents that inhibit the activation states of microglia and/or astroglia (including IL-10, minocycline, etanercept, propentofylline, ibudilast, licofelone, SP600125, carbenoxolone). Reversal of pain-related behavior has also been shown with disruption by an inhibitor (CR8) and/or genetic deletion of cell cycle-related proteins, deletion of a truncated receptor (trkB.T1) for brain-derived neurotrophic factor (BDNF), or reduction by antisense knockdown or an inhibitor (AMG9810) of the activity of channels (TRPV1 or Nav1.8) important for electrical activity in primary nociceptors. Nociceptor activity is known to drive central neuroinflammation in peripheral injury models, and nociceptors appear to be an integral component of host defense. Thus, emerging results suggest that spinal and systemic effects of SCI can activate nociceptor-mediated host defense responses that interact via neuroinflammatory signaling with complex central consequences of SCI to drive chronic pain. This broader view of SCI-induced neuroinflammation suggests new targets, and additional complications, for efforts to develop effective treatments for neuropathic SCI pain.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, University of Texas Medical School at Houston, TX, USA.
| |
Collapse
|
24
|
da Silva MD, Bobinski F, Sato KL, Kolker SJ, Sluka KA, Santos ARS. IL-10 cytokine released from M2 macrophages is crucial for analgesic and anti-inflammatory effects of acupuncture in a model of inflammatory muscle pain. Mol Neurobiol 2014; 51:19-31. [PMID: 24961568 DOI: 10.1007/s12035-014-8790-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/09/2014] [Indexed: 12/14/2022]
Abstract
Muscle pain is a common medical problem that is difficult to treat. One nonpharmacological treatment used is acupuncture, a procedure in which fine needles are inserted into body points with the intent of relieving pain and other symptoms. Here we investigated the effects of manual acupuncture (MA) on modulating macrophage phenotype and interleukin-10 (IL-10) concentrations in animals with muscle inflammation. Carrageenan, injected in the gastrocnemius muscle of mice, induces an inflammatory response characterized by mechanical hyperalgesia and edema. The inflammation is initially a neutrophilic infiltration that converts to a macrophage-dominated inflammation by 48 h. MA of the Sanyinjiao or Spleen 6 (SP6) acupoint reduces nociceptive behaviors, heat, and mechanical hyperalgesia and enhanced escape/avoidance and the accompanying edema. SP6 MA increased muscle IL-10 levels and was ineffective in reducing pain behaviors and edema in IL-10 knockout (IL-10(-/-)) mice. Repeated daily treatments with SP6 MA induced a phenotypic switch of muscle macrophages with reduced M1 macrophages (pro-inflammatory cells) and an increase of M2 macrophages (anti-inflammatory cells and important IL-10 source). These findings provide new evidence that MA produces a phenotypic switch in macrophages and increases IL-10 concentrations in muscle to reduce pain and inflammation.
Collapse
Affiliation(s)
- Morgana D da Silva
- Laboratório de Neurobiologia da Dor e Inflamação, Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, 88040-900, Brazil
| | | | | | | | | | | |
Collapse
|
25
|
Dengler EC, Alberti LA, Bowman BN, Kerwin AA, Wilkerson JL, Moezzi DR, Limanovich E, Wallace JA, Milligan ED. Improvement of spinal non-viral IL-10 gene delivery by D-mannose as a transgene adjuvant to control chronic neuropathic pain. J Neuroinflammation 2014; 11:92. [PMID: 24884664 PMCID: PMC4046049 DOI: 10.1186/1742-2094-11-92] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Peri-spinal subarachnoid (intrathecal; i.t.) injection of non-viral naked plasmid DNA encoding the anti-inflammatory cytokine, IL-10 (pDNA-IL-10) suppresses chronic neuropathic pain in animal models. However, two sequential i.t. pDNA injections are required within a discrete 5 to 72-hour period for prolonged efficacy. Previous reports identified phagocytic immune cells present in the peri-spinal milieu surrounding the i.t injection site that may play a role in transgene uptake resulting in subsequent IL-10 transgene expression. METHODS In the present study, we aimed to examine whether factors known to induce pro-phagocytic anti-inflammatory properties of immune cells improve i.t. IL-10 transgene uptake using reduced naked pDNA-IL-10 doses previously determined ineffective. Both the synthetic glucocorticoid, dexamethasone, and the hexose sugar, D-mannose, were factors examined that could optimize i.t. pDNA-IL-10 uptake leading to enduring suppression of neuropathic pain as assessed by light touch sensitivity of the rat hindpaw (allodynia). RESULTS Compared to dexamethasone, i.t. mannose pretreatment significantly and dose-dependently prolonged pDNA-IL-10 pain suppressive effects, reduced spinal IL-1β and enhanced spinal and dorsal root ganglia IL-10 immunoreactivity. Macrophages exposed to D-mannose revealed reduced proinflammatory TNF-α, IL-1β, and nitric oxide, and increased IL-10 protein release, while IL-4 revealed no improvement in transgene uptake. Separately, D-mannose dramatically increased pDNA-derived IL-10 protein release in culture supernatants. Lastly, a single i.t. co-injection of mannose with a 25-fold lower pDNA-IL-10 dose produced prolonged pain suppression in neuropathic rats. CONCLUSIONS Peri-spinal treatment with D-mannose may optimize naked pDNA-IL-10 transgene uptake for suppression of allodynia, and is a novel approach to tune spinal immune cells toward pro-phagocytic phenotype for improved non-viral gene therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Erin D Milligan
- Department of Neurosciences, UNM School of Medicine, University of New Mexico Health Sciences Center, 1 University of New Mexico, Albuquerque, NM 87131-0001, USA.
| |
Collapse
|
26
|
Thompson CD, Zurko JC, Hanna BF, Hellenbrand DJ, Hanna A. The therapeutic role of interleukin-10 after spinal cord injury. J Neurotrauma 2013; 30:1311-24. [PMID: 23731227 DOI: 10.1089/neu.2012.2651] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition affecting 270,000 people in the United States. A potential treatment for decreasing the secondary inflammation, excitotoxic damage, and neuronal apoptosis associated with SCI, is the anti-inflammatory cytokine interleukin-10. The best characterized effects of IL-10 are anti-inflammatory-it downregulates pro-inflammatory species interleukin-1β (IL-1β), interleukin-2 (IL-2), interleukin-6 (IL-6), tumor necrosis factor-α, interferon-γ, matrix metalloproteinase-9, nitric oxide synthase, myeloperoxidase, and reactive oxygen species. Pro-apoptotic factors cytochrome c, caspase 3, and Bax are downregulated by IL-10, whereas anti-apoptotic factors B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X, B-cell lymphoma-extra large (Bcl-xl) are upregulated by IL-10. IL-10 also provides trophic support to neurons through the IL-10 receptor. Increased tissue sparing, functional recovery, and neuroprotection are seen with an immediate post-SCI systemic administration of IL-10. Treatment of SCI with IL-10 has been used successfully in combination with Schwann cell and olfactory glial cell grafts, as well as methylprednisolone. Minocycline, tetramethylpyrazine, and hyperbaric oxygen treatment all increase IL-10 levels in a SCI models and result in increased tissue sparing and functional recovery. A chronic systemic administration of IL-10 does not appear to be beneficial to SCI recovery and causes increased susceptibility to septicemia, pneumonia, and peripheral neuropathy. However, a localized upregulation of IL-10 has been shown to be beneficial and can be achieved by herpes simplex virus gene therapy, injection of poliovirus replicons, or surgical placement of a slow-release compound. IL-10 shows promise as a treatment for SCI, although research on local IL-10 delivery timeline and dosage needs to be expanded.
Collapse
Affiliation(s)
- Colton D Thompson
- Department of Neurological Surgery, University of Wisconsin , Madison, Wisconsin, USA
| | | | | | | | | |
Collapse
|
27
|
Milligan ED, Penzkover KR, Soderquist RG, Mahoney MJ. Spinal interleukin-10 therapy to treat peripheral neuropathic pain. Neuromodulation 2012; 15:520-6; discussion 526. [PMID: 22672183 DOI: 10.1111/j.1525-1403.2012.00462.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Current research indicates that chronic peripheral neuropathic pain includes a role for glia and the actions of proinflammatory factors. This review briefly discusses the glial and cytokine responses that occur following peripheral nerve damage in support of utilizing anti-inflammatory cytokine interleukin-10 (IL-10) therapy to suppress chronic peripheral neuropathic pain. SPINAL NONVIRAL INTERLEUKIN-10 GENE THERAPY: IL-10 is one of the most powerful endogenous counter-regulators of proinflammatory cytokine function that acts in the nervous system. Subarachnoid (intrathecal) spinal injection of the gene encoding IL-10 delivered by nonviral vectors has several advantages over virally mediated gene transfer methods and leads to profound pain relief in several animal models. NONVIRAL GENE DELIVERY: Lastly, data are reviewed that nonviral deoxyribonucleic acid (DNA) encapsulated by a biologically safe copolymer, poly(lactic-co-glycolic) acid (PLGA), thought to protect DNA, leads to significantly improved therapeutic gene transfer in animal models, which additionally and significantly extends pain relief. CONCLUSIONS The impact of these early studies exploring anti-inflammatory genes emphasizes the exceptional therapeutic potential of new biocompatible intrathecal nonviral gene delivery approaches such as PLGA microparticles. Ultimately, ongoing expression of therapeutic genes is a viable option to treat chronic neuropathic pain in the clinic.
Collapse
Affiliation(s)
- Erin D Milligan
- Department of Neurosciences, University of New Mexico-Health Sciences Center, School of Medicine, NM, USA
| | | | | | | |
Collapse
|
28
|
Discovering cytokines as targets for chemotherapy-induced painful peripheral neuropathy. Cytokine 2012; 59:3-9. [PMID: 22537849 DOI: 10.1016/j.cyto.2012.03.027] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 03/07/2012] [Accepted: 03/29/2012] [Indexed: 11/23/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN), a dose-limiting neurotoxic effect of chemotherapy, is the most common reason for early cessation of cancer treatment. This can result in an increased risk of recurrence and decreased survival rate. Inflammatory cascade activation, proinflammatory cytokine upregulation, and neuro-immune communication pathways play essential roles in the initiation and progression of CIPN. Most notably, TNF-α, IL-1β, IL-6, and CCL2 are involved in neuropathic pain. Further elucidation of the role of these cytokines could lead to their development and use as biomarkers for predicting the onset of painful peripheral neuropathy and early axonal damage. In this review, we provide evidence for the involvement of cytokines in CIPN, the possible underlying mechanisms, and their use as potential therapeutic targets and biomarkers to prevent and improve the painful peripheral neuropathy related to chemotherapeutic agents.
Collapse
|
29
|
David S, López-Vales R, Wee Yong V. Harmful and beneficial effects of inflammation after spinal cord injury: potential therapeutic implications. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:485-502. [PMID: 23098732 DOI: 10.1016/b978-0-444-52137-8.00030-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Spinal cord injury (SCI) results in immediate damage followed by a secondary phase of tissue damage that occurs over a period of several weeks. The mechanisms underlying this secondary damage are multiple and not fully understood. A number of studies suggest that the local inflammatory response in the spinal cord that occurs after SCI contributes importantly to secondary damage. This response is mediated by cells normally found in the central nervous system (CNS) as well as infiltrating leukocytes. While the inflammatory response mediated by these cells is required for efficient clearance of tissue debris, and promotes wound healing and tissue repair, they also release various factors that can be detrimental to neurons, glia, axons, and myelin. In this chapter we provide an overview of the inflammatory response at the cell and molecular level after SCI, and review the current state of knowledge about its contribution to tissue damage and repair. Additionally, we discuss how some of this work is leading to the development and testing of drugs that modulate inflammation to treat acute SCI in humans.
Collapse
Affiliation(s)
- Samuel David
- McGill University Health Centre, Montreal, Canada.
| | | | | |
Collapse
|
30
|
Kiyota T, Ingraham KL, Swan RJ, Jacobsen MT, Andrews SJ, Ikezu T. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP+PS1 mice. Gene Ther 2011; 19:724-33. [PMID: 21918553 PMCID: PMC3241853 DOI: 10.1038/gt.2011.126] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brain inflammation is a double-edged sword: it is required for brain repair in acute damage, whereas chronic inflammation and autoimmune disorders are neuropathogenic. Certain pro-inflammatory cytokines and chemokines are closely related to cognitive dysfunction and neurodegeneration. Representative anti-inflammatory cytokines, such as interleukin (IL)-10, can suppress neuroinflammation and have significant therapeutic potentials in ameliorating neurodegenerative disorders, such as Alzheimer’s disease (AD). Here, we show that adeno-associated virus (AAV) serotype 2/1 hybrid-mediated neuronal expression of the mouse IL-10 gene ameliorates cognitive dysfunction in APP+PS1 bigenic mice. AAV2/1 infection of hippocampal neurons resulted in sustained expression of IL-10 without its leakage into the blood, reduced astro/microgliosis, enhanced plasma amyloid-β peptide (Aβ) levels, and enhanced neurogenesis. Moreover, increased levels of IL-10 improved spatial learning as determined by the radial arm water maze. Finally, IL-10-stimulated microglia enhanced proliferation but not differentiation of primary neural stem cells in the co-culture system, while IL-10 itself had no effect. Our data suggest that IL-10 gene delivery has a therapeutic potential for a non-Aβ-targeted treatment of AD.
Collapse
Affiliation(s)
- T Kiyota
- Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, USA
| | | | | | | | | | | |
Collapse
|
31
|
Gonzalez P, Peluffo H, Acarin L, Villaverde A, Gonzalez B, Castellano B. Interleukin-10 overexpression does not synergize with the neuroprotective action of RGD-containing vectors after postnatal brain excitotoxicity but modulates the main inflammatory cell responses. J Neurosci Res 2011; 90:143-59. [PMID: 21922521 DOI: 10.1002/jnr.22741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 06/05/2011] [Accepted: 06/16/2011] [Indexed: 11/07/2022]
Abstract
Antiinflammatory cytokines such as interleukin-10 (IL-10) have been used to modulate and terminate inflammation and provide neuroprotection. Recently, we reported that the modular recombinant transfection vector NLSCt is an efficient tool for transgene overexpression in vivo, which induces neuroprotection as a result of its RGD-mediated integrin-interacting capacity. We here sought to evaluate the putative synergic neuroprotective action exerted by IL-10 overexpression using NLSCt as a transfection vector after an excitotoxic injury to the postnatal rat brain. For this purpose, lesion volume, neurodegeneration, astroglial and microglial responses, neutrophil infiltration, and proinflammatory cytokine production were analyzed at several survival times after intracortical NMDA injection in postnatal day 9 rats, followed by injection of NLSCt combined with the IL-10 gene, a control transgene, or saline vehicle solution. Our results show no combined neuroprotective effect between RGD-interacting vectors and IL-10 gene therapy; instead, IL-10 overexpression using NLSCt as transfection vector increased lesion volume and neuronal degeneration at 12 hr and 3 days postlesion. In parallel, NLSCt/IL-10 treated animals displayed increased density of neutrophils and microglia/macrophages, and a reduced astroglial content of GFAP and vimentin. Moreover, NLSCt/IL-10 treated animals did not show any variation in interleukin-1β or tumor necrosis factor-α expression but a slight increase in interleukin-6 content at 7 days postlesion. In conclusion, overexpression of IL-10 by using NLSCt transfection vector did not synergistically neuroprotect the excitotoxically damaged postnatal rat brain but induced changes in the astroglial and microglial and inflammatory cell response.
Collapse
Affiliation(s)
- Pau Gonzalez
- Unit of Medical Histology, Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Autonomous University of Barcelona, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
32
|
Soderquist RG, Milligan ED, Harrison JA, Chavez RA, Johnson KW, Watkins LR, Mahoney MJ. PEGylation of interleukin-10 for the mitigation of enhanced pain states. J Biomed Mater Res A 2010; 93:1169-79. [PMID: 19768789 DOI: 10.1002/jbm.a.32611] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The anti-inflammatory cytokine interleukin-10 (IL-10) shows promise for the treatment of neuropathic pain, but for IL-10 to be clinically useful as a short-term therapeutic its duration needs to be improved. In this study, IL-10 was covalently modified with polyethylene glycol (PEG) with the goal of stabilizing and increasing protein levels in the CSF to improve the efficacy of IL-10 for treating neuropathic pain. Two different PEGylation methods were explored in vitro to identify suitable PEGylated IL-10 products for subsequent in vivo testing. PEGylation of IL-10 by acylation yielded a highly PEGylated product with a 35-fold in vitro biological activity reduction. PEGylation of IL-10 by reductive amination yielded products with a minimal number of PEG molecules attached and in vitro biological activity reductions of approximately 3-fold. In vivo collections of cerebrospinal fluid after intrathecal administration demonstrated that 20 kDa PEG attachment to IL-10 increased the concentration of IL-10 in the cerebrospinal fluid over time. Relative to unmodified IL-10, the 20 kDa PEG-IL-10 product exhibited an increased therapeutic duration and magnitude in an animal model of neuropathic pain. This suggests that PEGylation is a viable strategy for the short-term treatment or, in conjunction with other approaches, the long-term treatment of enhanced pain states.
Collapse
Affiliation(s)
- Ryan G Soderquist
- Department of Chemical & Biological Engineering, University of Colorado at Boulder, Boulder, Colorado 80309, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Hoschouer EL, Finseth T, Flinn S, Basso DM, Jakeman LB. Sensory stimulation prior to spinal cord injury induces post-injury dysesthesia in mice. J Neurotrauma 2010; 27:777-87. [PMID: 20121420 PMCID: PMC2943942 DOI: 10.1089/neu.2009.1182] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chronic pain and dysesthesias are debilitating conditions that can arise following spinal cord injury (SCI). Research studies frequently employ rodent models of SCI to better understand the underlying mechanisms and develop better treatments for these phenomena. While evoked withdrawal tests can assess hypersensitivity in these SCI models, there is little consensus over how to evaluate spontaneous sensory abnormalities that are seen in clinical SCI subjects. Overgrooming (OG) and biting after peripheral nerve injury or spinal cord excitotoxic lesions are thought to be one behavioral demonstration of spontaneous neuropathic pain or dysesthesia. However, reports of OG after contusion SCI are largely anecdotal and conditions causing this response are poorly understood. The present study investigated whether repeated application of sensory stimuli to the trunk prior to mid-thoracic contusion SCI would induce OG after SCI in mice. One week prior to SCI or laminectomy, mice were subjected either to nociceptive and mechanical stimulation, mechanical stimulation only, the testing situation without stimulation, or no treatment. They were then examined for 14 days after surgery and the sizes and locations of OG sites were recorded on anatomical maps. Mice subjected to either stimulus paradigm showed increased OG compared with unstimulated or uninjured mice. Histological analysis showed no difference in spinal cord lesion size due to sensory stimulation, or between mice that overgroomed or did not overgroom. The relationship between prior stimulation and contusion injury in mice that display OG indicates a critical interaction that may underlie one facet of spontaneous neuropathic symptoms after SCI.
Collapse
Affiliation(s)
- Emily L. Hoschouer
- Department of Physiology and Cell Biology, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Neuroscience Graduate Studies Program, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Center for Brain and Spinal Cord Repair, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Taylor Finseth
- College of Medicine, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Sharon Flinn
- Division of Occupational Therapy, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - D. Michele Basso
- Department of Neuroscience, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Neuroscience Graduate Studies Program, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Center for Brain and Spinal Cord Repair, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Division of Physical Therapy, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
| | - Lyn B. Jakeman
- Department of Physiology and Cell Biology, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Department of Neuroscience, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Neuroscience Graduate Studies Program, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
- Center for Brain and Spinal Cord Repair, School of Allied Medicine, The Ohio State University Medical Center, Columbus, Ohio
| |
Collapse
|
34
|
Shimizu K, Guo W, Wang H, Zou S, LaGraize SC, Iwata K, Wei F, Dubner R, Ren K. Differential involvement of trigeminal transition zone and laminated subnucleus caudalis in orofacial deep and cutaneous hyperalgesia: the effects of interleukin-10 and glial inhibitors. Mol Pain 2009; 5:75. [PMID: 20025765 PMCID: PMC2806354 DOI: 10.1186/1744-8069-5-75] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 12/21/2009] [Indexed: 01/11/2023] Open
Abstract
Background In addition to caudal subnucleus caudalis (Vc) of the spinal trigeminal complex, recent studies indicate that the subnuclei interpolaris/caudalis (Vi/Vc) transition zone plays a unique role in processing deep orofacial nociceptive input. Studies also suggest that glia and inflammatory cytokines contribute to the development of persistent pain. By systematically comparing the effects of microinjection of the antiinflammatory cytokine interleukin (IL)-10 and two glial inhibitors, fluorocitrate and minocycline, we tested the hypothesis that there was a differential involvement of Vi/Vc and caudal Vc structures in deep and cutaneous orofacial pain. Results Deep or cutaneous inflammatory hyperalgesia, assessed with von Frey filaments, was induced in rats by injecting complete Freund's adjuvant (CFA) into the masseter muscle or skin overlying the masseter, respectively. A unilateral injection of CFA into the masseter or skin induced ipsilateral hyperalgesia that started at 30 min, peaked at 1 d and lasted for 1-2 weeks. Secondary hyperalgesia on the contralateral site also developed in masseter-, but not skin-inflamed rats. Focal microinjection of IL-10 (0.006-1 ng), fluorocitrate (1 μg), and minocycline (0.1-1 μg) into the ventral Vi/Vc significantly attenuated masseter hyperalgesia bilaterally but without an effect on hyperalgesia after cutaneous inflammation. Injection of the same doses of these agents into the caudal Vc attenuated ipsilateral hyperalgesia after masseter and skin inflammation, but had no effect on contralateral hyperalgesia after masseter inflammation. Injection of CFA into the masseter produced significant increases in N-methyl-D-aspartate (NMDA) receptor NR1 serine 896 phosphorylation and glial fibrillary acidic protein (GFAP) levels, a marker of reactive astrocytes, in Vi/Vc and caudal Vc. In contrast, cutaneous inflammation only produced similar increases in the Vc. Conclusion These results support the hypothesis that the Vi/Vc transition zone is involved in deep orofacial injury and suggest that glial inhibition and interruption of the cytokine cascade after inflammation may provide pain relief.
Collapse
Affiliation(s)
- Kohei Shimizu
- Department of Neural and Pain Sciences, Dental School, University of Maryland, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Reyes-Gibby CC, Spitz MR, Yennurajalingam S, Swartz M, Gu J, Wu X, Bruera E, Shete S. Role of inflammation gene polymorphisms on pain severity in lung cancer patients. Cancer Epidemiol Biomarkers Prev 2009; 18:2636-42. [PMID: 19773451 DOI: 10.1158/1055-9965.epi-09-0426] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Many of the same inflammatory factors that promote tumor growth are also hypothesized to function as pain modulators. There is substantial interindividual variation in pain severity in cancer patients. Therefore, we evaluated 59 single nucleotide polymorphisms in 37 inflammation genes in newly diagnosed non-Hispanic Caucasian lung cancer patients (n = 667) and assessed their association with pain severity. Patients rated their pain "during the past week" on an 11-point numeric scale (0 = "no pain" and 10 = "pain as bad as you can imagine") at presentation before initiating cancer therapy. Reported analgesic use was abstracted from charts and converted to morphine equivalent daily dose. Results showed that 16% of the patients reported severe pain (score > or = 7). Advanced stage of disease [odds ratio (OR), 2.34; 95% confidence interval (95% CI), 1.50-3.65; P = 0.001], age < or = 50 years (OR, 2.10; 95% CI, 1.32-3.30; P = 0.002), reports of depressed mood (OR, 3.68; 95% CI, 1.96-6.93; P = 0.001), fatigue (OR, 3.72; 95% CI, 2.36-5.87; P = 0.001), and morphine equivalent daily dose (OR, 1.02; 95% CI, 1.01-1.03) were significantly correlated with severe pain. Controlling for these nongenetic covariates, we found that patients with CC genotypes for PTGS2 exon10+837T>C (rs5275) were at lower risk for severe pain (OR, 0.33; 95% CI, 0.11-0.97) and an additive model for TNFalpha -308GA (rs1800629; OR, 1.67; 95% CI, 1.08-2.58) and NFKBIA Ex6+50C>T (rs8904) was predictive of severe pain (OR, 0.64; 95% CI, 0.43-0.93). In a multigene analysis, we found a gene-dose effect, with each protective genotype reducing the risk for severe pain by as much as 38%. This study suggests the importance of inflammation gene polymorphisms in modulating pain severity. Additional studies are needed to validate our findings.
Collapse
Affiliation(s)
- Cielito C Reyes-Gibby
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, Unit 1340, 1155 Pressler Street, Houston, TX 77030-4009, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhou Z, Peng X, Insolera R, Fink DJ, Mata M. IL-10 promotes neuronal survival following spinal cord injury. Exp Neurol 2009; 220:183-90. [PMID: 19716366 DOI: 10.1016/j.expneurol.2009.08.018] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 08/18/2009] [Accepted: 08/18/2009] [Indexed: 11/29/2022]
Abstract
We have previously reported that the anti-inflammatory cytokine IL-10 induces a number of signaling cascades through the IL-10 receptor in spinal cord neurons in vitro to activate NF-kappaB transcription Bcl-2 and Bcl-x(L) and that, after exposure to glutamate IL-10, blocks cytochrome c release and caspase cleavage. In the current study we used a herpes simplex virus (HSV)-based vector to express IL-10 in spinal cord in vivo. Injection of the vector 30 minutes after lateral hemisection injury resulted in increased neuronal survival in the anterior quadrant of the spinal cord and improved motor function up to 6 weeks after injury, that correlated with translocation of p50 and p65 NF-kappaB to the nucleus and increased expression of Bcl-2 and Bcl-x(L) in anterior quadrant neurons. Inhibition of cytochrome c release and caspase 3 cleavage was seen in homogenates of injured spinal cord treated by the IL-10 vector. Taken together with in vitro studies that demonstrate direct neuroprotective effects of IL-10 acting through the neuronal IL-10 receptor, these results suggest that IL-10 may provide direct neuroprotective effects in spinal cord injury separate from and in addition to the known anti-inflammatory effects and point to the possibility that IL-10 delivery by gene transfer may be a useful adjunctive therapy for spinal cord injury.
Collapse
Affiliation(s)
- Zhigang Zhou
- Department of Neurology, University of Michigan and Ann Arbor VA Healthcare System, 5027 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
37
|
Eisener-Dorman AF, Lawrence DA, Bolivar VJ. Cautionary insights on knockout mouse studies: the gene or not the gene? Brain Behav Immun 2009; 23:318-24. [PMID: 18822367 PMCID: PMC2746382 DOI: 10.1016/j.bbi.2008.09.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 09/02/2008] [Accepted: 09/03/2008] [Indexed: 01/03/2023] Open
Abstract
Gene modification technologies play a vital role in the study of biological systems and pathways. Although there is widespread and beneficial use of genetic mouse models, potential shortcomings of gene targeting technology exist, and are not always taken into consideration. Oversights associated with the technology can lead to misinterpretation of results; for example, ablation of a gene of interest can appear to cause an observed phenotype when, in fact, residual embryonic stem cell-derived genetic material in the genetic background or in the area immediately surrounding the ablated gene is actually responsible. The purpose of this review is to remind researchers, regardless of scientific discipline, that the background genetics of a knockout strain can have a profound influence on any observed phenotype. It is important that this issue be appropriately addressed during data collection and interpretation.
Collapse
Affiliation(s)
- Amy F. Eisener-Dorman
- Wadsworth Center, 120 New Scotland Avenue, Albany, New York 12208, USA, School of Public Health, Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, 12201, USA
| | - David A. Lawrence
- Wadsworth Center, 120 New Scotland Avenue, Albany, New York 12208, USA, School of Public Health, Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, 12201, USA
| | - Valerie J. Bolivar
- Wadsworth Center, 120 New Scotland Avenue, Albany, New York 12208, USA, School of Public Health, Department of Biomedical Sciences, State University of New York at Albany, Albany, New York, 12201, USA
| |
Collapse
|
38
|
Milligan ED, Sloane EM, Watkins LR. Glia in pathological pain: a role for fractalkine. J Neuroimmunol 2008; 198:113-20. [PMID: 18547654 PMCID: PMC3289257 DOI: 10.1016/j.jneuroim.2008.04.011] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 04/10/2008] [Indexed: 01/01/2023]
Abstract
Microglia and/or astrocytes play a significant role in the creation and maintenance of exaggerated pain states with inflammatory and/or neuropathic etiologies. The chemokine, fractalkine, has several functions, including the newly recognized role of mediating neuropathic pain conditions. Although constitutively expressed and released during inflammation, increased release of fractalkine binds to and activates microglia leading to pathological pain. We review the critical role of fractalkine in neuron-to-glial communication after peripheral nerve injury and inflammation and explore anti-inflammatory cytokines like interleukin-10 as a novel and effective approach for clinical pain control.
Collapse
Affiliation(s)
- E D Milligan
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM 87131-0001 USA.
| | | | | |
Collapse
|
39
|
Levin SG, Godukhin OV. Protective effects of interleukin-10 on the development of epileptiform activity evoked by transient episodes of hypoxia in rat hippocampal slices. ACTA ACUST UNITED AC 2007; 37:467-70. [PMID: 17505796 DOI: 10.1007/s11055-007-0036-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2005] [Accepted: 12/12/2005] [Indexed: 11/24/2022]
Abstract
The aim of the present work was to study the effects of interleukin-10 at concentrations of 1 and 10 ng/ml on the development of epileptiform discharges evoked in pyramidal neurons in field CA3 in rat hippocampal slices by transient episodes of hypoxia. Three 3-min episodes of hypoxia led to decreases in the generation threshold for evoked trains of population spikes and an increase in the number of population spikes per train in pyramidal neurons of field CA1. Interleukin-10 at a concentration of 1 ng/ml completely eliminated the development of epileptiform activity, while its protective effect was less marked at a concentration of 10 ng/ml. These effects of interleukin-10 on living hippocampal slices in in vitro conditions show that they may be associated with the functions of this cytokine as an intercellular mediator of the central nervous system itself rather than being mediated by the peripheral immune system. The results of these studies provide the first experimental evidence of the action of the anti-inflammatory cytokine interleukin-10 on the development of hypoxia-evoked epileptiform events in the hippocampus.
Collapse
Affiliation(s)
- S G Levin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia.
| | | |
Collapse
|
40
|
Brewer KL, Nolan TA. Spinal and supraspinal changes in tumor necrosis factor-alpha expression following excitotoxic spinal cord injury. J Mol Neurosci 2007; 31:13-21. [PMID: 17416966 DOI: 10.1007/bf02686114] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Revised: 05/12/2006] [Accepted: 05/12/2006] [Indexed: 12/22/2022]
Abstract
The role of tumor necrosis factor-alpha (TNF-alpha) after spinal cord injury (SCI) is well characterized in the cord, but the impact of this inflammatory process on supraspinal levels is unknown. This study examines TNF-alpha mRNA and protein levels in the brains and spinal cords of mice after SCI. Mice received intraspinal injections of quisqualic acid (QUIS) to create an excitotoxic injury that is known to result in pain behaviors. An ELISA determined serum levels of TNF-alpha, whereas real-time PCR and Western blot analysis were used to determine mRNA and protein levels, respectively, at 3, 6, 12, 24, 48, 72 h, or 14 d postinjury. No difference existed in serum TNF-alpha levels between sham- and QUIS-injected animals. TNF-alpha mRNA in the cord was increased at 3, 6, 12, and 24 h in QUIS-injected animals relative to shams. TNF-alpha protein was elevated at 12 and 48 h postinjury. TNF-alpha mRNA levels in the brain were elevated at 12 and 24 h, with elevated protein levels at 6 h. Animals that developed pain behaviors had increased levels of TNF-alpha mRNA in the brain. Excitotoxic SCI results in altered TNF-alpha mRNA and protein levels in the cords and brains of mice within 6 h of injury. These changes likely contribute to the pathogenesis of injury within the cord. The role of TNF-alpha in the brain postinjury has not been defined but might contribute to the development of pain post-SCI.
Collapse
Affiliation(s)
- Kori L Brewer
- Department of Emergency Medicine, PCMH 3ED304, East Carolina University Brody School of Medicine, Greenville, NC 27834, USA.
| | | |
Collapse
|
41
|
Kelly MC, Kosloff BR, Morrow CD. Forced selection of tRNA(Glu) reveals the importance of two adenosine-rich RNA loops within the U5-PBS for SIV(smmPBj) replication. Virology 2007; 366:330-9. [PMID: 17543363 PMCID: PMC2040306 DOI: 10.1016/j.virol.2007.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 03/15/2007] [Accepted: 04/25/2007] [Indexed: 11/23/2022]
Abstract
Simian immunodeficiency virus (SIV) and human immunodeficiency virus (HIV-1) preferentially select and use tRNA(Lys,3) as the primer for initiation of reverse transcription. Previous studies have shown that HIV-1 can be forced to use tRNA(Glu) if mutations are made within the primer-binding site (PBS) and a region upstream, A-loop, to be complementary to the 3'-terminal 18 nucleotides and anticodon loop of tRNA(Glu). To examine the primer preference of SIV, mutations were made within the PBS of SIV(smmPBj) to be complementary to tRNA(Glu). Analysis of the production of infectious virus revealed that SIV(smmPBj) with the PBS complementary to tRNA(Glu) retained approximately 80% infectivity of the wild type. However, modification of the U5 of SIV(smmPBj) to alter nucleotides to be complementary to the anticodon of tRNA(Glu), in combination with the PBS complementary to tRNA(Glu), drastically reduced the production of infectious SIV(smmPBj) to less than 1% that of wild type. The replication of SIV(smmPBj) with the PBS complementary to tRNA(Glu) was similar to that of the wild type virus, while the replication of SIV(smmPBj) with PBS and A-loop complementary to tRNA(Glu) was delayed compared to that of wild type virus. Analysis of the PBS regions revealed that the virus with the PBS complementary to tRNA(Glu) reverted quickly, within 4 days, to be complementary to tRNA(Lys,3), while the virus with PBS and A-loop complementary to tRNA(Glu) retained the PBS for a longer time during in vitro culture although following extended replication both the A-loop and PBS of SIV(smmPBj) reverted to be complementary to tRNA(Lys,3). RNA modeling of SIV(smmPBj) U5-PBS by m-fold revealed two potential A-loop regions. Mutations in either A-loop drastically effected replication in human PBMC. Analysis of the A-loops following in vitro replication revealed that both reverted to the wild type sequence. The results of these studies demonstrate that SIV(smmPBj), like HIV-1, preferentially utilizes tRNA(Lys,3) as a primer for reverse transcription for high level replication, but unlike HIV-1 selection may involve the use of two adenosine-rich loops.
Collapse
Affiliation(s)
| | | | - Casey D. Morrow
- * Corresponding author: Casey D. Morrow, Department of Cell Biology, University of Alabama at Birmingham, 802 Kaul Building, 720 20 Street South, Birmingham, Alabama 35294 (USA), 205 934 5705 (Phone), 205 934 1580 (FAX),
| |
Collapse
|
42
|
|
43
|
Hendricks WA, Pak ES, Owensby JP, Menta KJ, Glazova M, Moretto J, Hollis S, Brewer KL, Murashov AK. Predifferentiated embryonic stem cells prevent chronic pain behaviors and restore sensory function following spinal cord injury in mice. Mol Med 2006; 12:34-46. [PMID: 16838066 PMCID: PMC1514553 DOI: 10.2119/2006-00014.hendricks] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Accepted: 03/25/2006] [Indexed: 11/06/2022] Open
Abstract
Embryonic stem (ES) cells have been investigated in repair of the CNS following neuronal injury and disease; however, the efficacy of these cells in treatment of postinjury pain is far from clear. In this study, we evaluated the therapeutic potential of predifferentiated mouse ES cells to restore sensory deficits following spinal cord injury (SCI) in mice. The pain model used unilateral intraspinal injection of quisqualic acid (QUIS) into the dorsal horn between vertebral levels T13 and L1. Seven days later, 60,000 predifferentiated ES cells or media were transplanted into the site of the lesion. Histological analysis at 7, 14, and 60 days post-transplantation revealed that animals receiving ES cell transplants suffered significantly less tissue damage than animals receiving media alone. Transplanted cells provided immediate effects on both spontaneous and evoked pain behaviors. Treatment with ES cells resulted in 0% (n = 28) excessive grooming behavior versus 60% (18 of 30) in media-treated animals. In the acetone test (to assess thermal allodynia), mice recovered to preinjury levels by 12 days after ES cell transplant, whereas control animals injected with media after SCI did not show any improvement up to 60 days. Similarly, the von Frey test (to assess mechanical allodynia) and the formalin test (to assess nociceptive hyperalgesia) showed that transplantation of predifferentiated ES cells significantly reduced these pain behaviors following injury. Here we show that predifferentiated ES cells act in a neuroprotective manner and provide antinociceptive and therapeutic effects following excitotoxic SCI.
Collapse
Affiliation(s)
- Wesley A Hendricks
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
- Department of Biology, East Carolina University, Greenville, NC 27834, USA
| | - Elena S Pak
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
| | - J Paul Owensby
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
- Department of Biology, East Carolina University, Greenville, NC 27834, USA
| | - Kristie J Menta
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
| | - Margarita Glazova
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
| | - Justin Moretto
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
| | - Sarah Hollis
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
| | - Kori L Brewer
- Department of Emergency Medicine, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
| | - Alexander K Murashov
- Department of Physiology, The Brody School of Medicine, East Carolina
University, Greenville, NC, USA
- Address correspondence and reprint requests to Alexander K. Murashov, East
Carolina University School of Medicine, Brody Bldg #6N-98, 600 Moye
Blvd, Greenville, NC 27834. Phone: 252-744-3111; fax: 252-744-3460; e-mail: ; web site: http://www.ecu.edu/physio/labakm
| |
Collapse
|
44
|
Kadiu I, Glanzer JG, Kipnis J, Gendelman HE, Thomas MP. Mononuclear phagocytes in the pathogenesis of neurodegenerative diseases. Neurotox Res 2006; 8:25-50. [PMID: 16260384 DOI: 10.1007/bf03033818] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Brain mononuclear phagocytes (MP, bone marrow monocyte-derived macrophages, perivascular macrophages, and microglia) function to protect the nervous system by acting as debris scavengers, killers of microbial pathogens, and regulators of immune responses. MP are activated by a variety of environmental cues and such inflammatory responses elicit cell injury and death in the nervous system. MP immunoregulatory responses include secretion of neurotoxic factors, mobilization of adaptive immunity, and cell chemotaxis. This incites tissue remodelling and blood-brain barrier dysfunction. As disease progresses, MP secretions engage neighboring cells in a vicious cycle of autocrine and paracrine amplification of inflammation leading to tissue injury and ultimately destruction. Such pathogenic processes tilt the balance between the relative production of neurotrophic and neurotoxic factors and to disease progression. The ultimate effects that brain MP play in disease revolves "principally" around their roles in neurodegeneration. Importantly, common functions of brain MP in neuroimmunity link highly divergent diseases (for example, human immunodeficiency virus type-one associated dementia, Alzheimer's disease and Parkinson's disease). Research into this process from our own laboratories and those of others seek to harness MP inflammatory processes with the intent of developing therapeutic interventions that block neurodegenerative processes and improve the quality of life in affected people.
Collapse
Affiliation(s)
- I Kadiu
- Laboratory of Neuroregeneration, Department of Pharmacology and Experimental Neuroscience, Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | | | |
Collapse
|
45
|
Wu Z, Zhang J, Nakanishi H. Leptomeningeal cells activate microglia and astrocytes to induce IL-10 production by releasing pro-inflammatory cytokines during systemic inflammation. J Neuroimmunol 2005; 167:90-8. [PMID: 16095726 DOI: 10.1016/j.jneuroim.2005.06.025] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Accepted: 06/20/2005] [Indexed: 11/29/2022]
Abstract
The leptomeninges covering the surface of the brain parenchyma play the physical role at the cerebrospinal fluid-blood barrier. We report here that leptomeningeal cells may transduce peripheral proinflammatory signals to the central anti-inflammatory response through the activation of glial cells in the brain parenchyma. After adjuvant injection, both microglia and astrocytes in the cerebral cortex localized in the proximity of the leptomeninges were activated. The protein levels of tumor necrosis factor-alpha (TNF-alpha) and interleukin-10 (IL-10) in the cortical extracts were significantly increased at different time after adjuvant injection. The TNF-alpha immunoreactivity was most prominent in the leptomeninges covering astrocytes. On the other hand, the IL-10 immunoreactivity was observed in both activated microglia and astrocytes localized along the leptomeninges. Cultured leptomeningeal cells covering the cerebral cortex released TNF-alpha which was significantly increased by lipopolysaccharide (LPS). Upon stimulation with LPS, cultured leptomeningeal cells also secreted interleukin-1beta and interleukin-6 with differential time-courses. When primary cultured rat astrocytes and microglia were treated with the conditioned medium of LPS-activated cultured leptomeningeal cells, the immunoreactivity of IL-10 was markedly increased. These observations strongly suggest that leptomeningeal cells release pro-inflammatory cytokines to activate both microglia and astrocytes during systemic inflammation. The activated astrocytes and microglia may in turn regulate anti-inflammatory response in the brain by providing IL-10.
Collapse
Affiliation(s)
- Zhou Wu
- Laboratory of Oral Aging Science, Faculty of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | |
Collapse
|
46
|
Cunningham MG, Donalds RA, Scouten CW, Tresch MC. A versatile, low-cost adaptor for stereotaxic and electrophysiologic spinal preparations in juvenile and adult rodents. Brain Res Bull 2005; 68:157-62. [PMID: 16325015 DOI: 10.1016/j.brainresbull.2005.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2005] [Revised: 07/19/2005] [Accepted: 08/08/2005] [Indexed: 11/23/2022]
Abstract
Rats and mice provide excellent models for normal spinal cord physiology, traumatic spinal cord injury, and various disease states. Alternative and improved methodologies for experimental spinal preparations are desirable, particularly in the wake of expanding neuroscience technology, such as the diverse array of transgenic mice now available, and exciting new therapeutic approaches, including transplantation and gene therapy. This report describes a simple, low-cost instrument for spinal preparations in rodents of different sizes, including rat pups. The device adapts to standard small animal stereotaxic instruments, precluding the need for additional stereotaxic apparatus. Surgical methods utilizing the device are presented demonstrating the instrument's capacity for precise alignment and stabilization of the spinal column that is reproducible from animal to animal. Proof of concept is demonstrated with results from spinal cord injections and electrophysiologic recordings.
Collapse
Affiliation(s)
- Miles G Cunningham
- Laboratory for Neural Reconstruction, McLean Hospital, Program in Neuroscience and Department of Psychiatry, Harvard Medical School, Boston, MA 02478, USA.
| | | | | | | |
Collapse
|
47
|
Abstract
During the past two decades, an important focus of pain research has been the study of chronic pain mechanisms, particularly the processes that lead to the abnormal sensitivity - spontaneous pain and hyperalgesia - that is associated with these states. For some time it has been recognized that inflammatory mediators released from immune cells can contribute to these persistent pain states. However, it has only recently become clear that immune cell products might have a crucial role not just in inflammatory pain, but also in neuropathic pain caused by damage to peripheral nerves or to the CNS.
Collapse
Affiliation(s)
- Fabien Marchand
- Neurorestoration Group and London Pain Consortium, Wolfson Wing, Hodgkin Building, Guy's Campus, King's College London, London Bridge, London SE1 1UL, UK
| | | | | |
Collapse
|
48
|
Milligan ED, Langer SJ, Sloane EM, He L, Wieseler-Frank J, O'Connor K, Martin D, Forsayeth JR, Maier SF, Johnson K, Chavez RA, Leinwand LA, Watkins LR. Controlling pathological pain by adenovirally driven spinal production of the anti-inflammatory cytokine, interleukin-10. Eur J Neurosci 2005; 21:2136-48. [PMID: 15869510 DOI: 10.1111/j.1460-9568.2005.04057.x] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gene therapy for the control of pain has, to date, targeted neurons. However, recent evidence supports that spinal cord glia are critical to the creation and maintenance of pain facilitation through the release of proinflammatory cytokines. Because of the ability of interleukin-10 (IL-10) to suppress proinflammatory cytokines, we tested whether an adenoviral vector encoding human IL-10 (AD-h-IL10) would block and reverse pain facilitation. Three pain models were examined, all of which are mediated by spinal pro-inflammatory cytokines. Acute intrathecal administration of rat IL-10 protein itself briefly reversed chronic constriction injury-induced mechanical allodynia and thermal hyperalgesia. The transient reversal caused by IL-10 protein paralleled the half-life of human IL-10 protein in the intrathecal space (t(1/2) approximately 2 h). IL-10 gene therapy both prevented and reversed thermal hyperalgesia and mechanical allodynia, without affecting basal responses to thermal or mechanical stimuli. Extra-territorial, as well as territorial, pain changes were reversed by this treatment. Intrathecal AD-h-IL10 injected over lumbosacral spinal cord led to elevated lumbosacral cerebrospinal fluid (CSF) levels of human IL-10, with far less human IL-10 observed in cervical CSF. In keeping with IL-10's known anti-inflammatory actions, AD-h-IL10 lowered CSF levels of IL-1, relative to control AD. These studies support that this gene therapy approach provides an alternative to neuronally focused drug and gene therapies for clinical pain control.
Collapse
Affiliation(s)
- Erin D Milligan
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jackson CA, Messinger J, Peduzzi JD, Ansardi DC, Morrow CD. Enhanced functional recovery from spinal cord injury following intrathecal or intramuscular administration of poliovirus replicons encoding IL-10. Virology 2005; 336:173-83. [DOI: 10.1016/j.virol.2005.03.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 03/03/2005] [Accepted: 03/15/2005] [Indexed: 11/26/2022]
|
50
|
Milligan ED, Sloane EM, Langer SJ, Cruz PE, Chacur M, Spataro L, Wieseler-Frank J, Hammack SE, Maier SF, Flotte TR, Forsayeth JR, Leinwand LA, Chavez R, Watkins LR. Controlling neuropathic pain by adeno-associated virus driven production of the anti-inflammatory cytokine, interleukin-10. Mol Pain 2005; 1:9. [PMID: 15813997 PMCID: PMC1079940 DOI: 10.1186/1744-8069-1-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2005] [Accepted: 02/25/2005] [Indexed: 01/06/2023] Open
Abstract
Despite many decades of drug development, effective therapies for neuropathic pain remain elusive. The recent recognition of spinal cord glia and glial pro-inflammatory cytokines as important contributors to neuropathic pain suggests an alternative therapeutic strategy; that is, targeting glial activation or its downstream consequences. While several glial-selective drugs have been successful in controlling neuropathic pain in animal models, none are optimal for human use. Thus the aim of the present studies was to explore a novel approach for controlling neuropathic pain. Here, an adeno-associated viral (serotype II; AAV2) vector was created that encodes the anti-inflammatory cytokine, interleukin-10 (IL-10). This anti-inflammatory cytokine is known to suppress the production of pro-inflammatory cytokines. Upon intrathecal administration, this novel AAV2-IL-10 vector was successful in transiently preventing and reversing neuropathic pain. Intrathecal administration of an AAV2 vector encoding beta-galactosidase revealed that AAV2 preferentially infects meningeal cells surrounding the CSF space. Taken together, these data provide initial support that intrathecal gene therapy to drive the production of IL-10 may prove to be an efficacious treatment for neuropathic pain.
Collapse
Affiliation(s)
- Erin D Milligan
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Evan M Sloane
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Stephen J Langer
- Department of Molecular, Cellular & Developmental Biology, University of CO at Boulder, Boulder, CO 80309 USA
| | - Pedro E Cruz
- Genetics Institute, the Powell Gene Therapy Center & Department of Pediatrics, University of FL at Gainesville, Gainesville, FL 32610 USA
| | - Marucia Chacur
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Leah Spataro
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Julie Wieseler-Frank
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Sayamwong E Hammack
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Steven F Maier
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| | - Terence R Flotte
- Genetics Institute, the Powell Gene Therapy Center & Department of Pediatrics, University of FL at Gainesville, Gainesville, FL 32610 USA
| | | | - Leslie A Leinwand
- Department of Molecular, Cellular & Developmental Biology, University of CO at Boulder, Boulder, CO 80309 USA
| | | | - Linda R Watkins
- Department of Psychology & the Center for Neuroscience, University of CO at Boulder, Boulder, CO 80309 USA
| |
Collapse
|