1
|
Silveira Prudente A, Hoon Lee S, Roh J, Luckemeyer DD, Cohen CF, Pertin M, Park CK, Suter MR, Decosterd I, Zhang JM, Ji RR, Berta T. Microglial STING activation alleviates nerve injury-induced neuropathic pain in male but not female mice. Brain Behav Immun 2024; 117:51-65. [PMID: 38190983 PMCID: PMC11034751 DOI: 10.1016/j.bbi.2024.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024] Open
Abstract
Microglia, resident immune cells in the central nervous system, play a role in neuroinflammation and the development of neuropathic pain. We found that the stimulator of interferon genes (STING) is predominantly expressed in spinal microglia and upregulated after peripheral nerve injury. However, mechanical allodynia, as a marker of neuropathic pain following peripheral nerve injury, did not require microglial STING expression. In contrast, STING activation by specific agonists (ADU-S100, 35 nmol) significantly alleviated neuropathic pain in male mice, but not female mice. STING activation in female mice leads to increase in proinflammatory cytokines that may counteract the analgesic effect of ADU-S100. Microglial STING expression and type I interferon-ß (IFN-ß) signaling were required for the analgesic effects of STING agonists in male mice. Mechanistically, downstream activation of TANK-binding kinase 1 (TBK1) and the production of IFN-ß, may partly account for the analgesic effect observed. These findings suggest that STING activation in spinal microglia could be a potential therapeutic intervention for neuropathic pain, particularly in males.
Collapse
Affiliation(s)
- Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Jueun Roh
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA; Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| | - Debora D Luckemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Cinder F Cohen
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Marie Pertin
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| | - Marc R Suter
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Franco-Enzástiga Ú, Natarajan K, David ET, Patel K, Ravirala A, Price TJ. Vinorelbine causes a neuropathic pain-like state in mice via STING and MNK1 signaling associated with type I interferon induction. iScience 2024; 27:108808. [PMID: 38303713 PMCID: PMC10831286 DOI: 10.1016/j.isci.2024.108808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/14/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Type I interferons (IFNs) increase the excitability of dorsal root ganglia (DRGs) neurons via MNK-eIF4E signaling to promote pain sensitization in mice. Activation of stimulator of interferon response cGAMP interactor 1 (STING) signaling is pivotal for type I IFN induction. We hypothesized that vinorelbine, a chemotherapeutic and activator of STING, would cause a neuropathic pain-like state in mice via STING signaling in DRG neurons associated with IFN production. Vinorelbine caused tactile allodynia and grimacing in wild-type (WT) mice and increased p-IRF3, type I IFNs, and p-eIF4E in peripheral nerves. Supporting our hypothesis, vinorelbine failed to induce IRF3-IFNs-MNK-eIF4E in StingGt/Gt mice and, subsequently, failed to cause pain. The vinorelbine-elicited increase of p-eIF4E was not observed in Mknk1-/- (MNK1 knockout) mice in peripheral nerves consistent with the attenuated pro-nociceptive effect of vinorelbine in these mice. Our findings show that activation of STING signaling in the periphery causes a neuropathic pain-like state through type I IFN signaling to DRG nociceptors.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Eric T. David
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Krish Patel
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Abhira Ravirala
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
3
|
Ma L, Deng D, Zhang T, Zhao W, Liu C, Huang S, Xu F, Wang Y, Zhao S, Ding Y, Huang Y, Wang K, Zhang Y, Yang X, Cao S, Chen X. STING-IFN-I pathway relieves incision induced acute postoperative pain via inhibiting the neuroinflammation in dorsal root ganglion of rats. Inflamm Res 2023; 72:1551-1565. [PMID: 37433890 DOI: 10.1007/s00011-023-01764-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND The purpose of this study was to study the effect of STING-IFN-I pathway on incision induced postoperative pain in rats and its possible mechanisms. METHODS The pain thresholds were evaluated by measuring the mechanical withdrawal threshold and the thermal withdrawal latency. The satellite glial cell and macrophage of DRG were analyzed. The expression of STING, IFN-a, P-P65, iNOS, TNF-α, IL-1β and IL-6 in DRG was evaluated. RESULTS The activation of STING-IFN-I pathway can reduce the mechanical hyperalgesia, thermal hyperalgesia, down-regulate the expression of P-P65, iNOS, TNF-α, IL-1β and IL-6, and inhibit the activation of satellite glial cell and macrophage in DRG. CONCLUSIONS The activation of STING-IFN-I pathway can alleviate incision induced acute postoperative pain by inhibiting the activation of satellite glial cell and macrophage, which reducing the corresponding neuroinflammation in DRG.
Collapse
Affiliation(s)
- Lulin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wenjing Zhao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chengxi Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Feng Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shuai Zhao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yan Huang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Anesthesiology, The First People's Hospital of Jiangxia District, Wuhan, China
| | - Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanyan Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinxin Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Song Cao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
Franco-Enzástiga Ú, Natarajan K, David ET, Patel KJ, Ravirala A, Price TJ. Vinorelbine causes a neuropathic pain-like state in mice via STING and MNK1 signaling associated with type I interferon induction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.03.543579. [PMID: 37333411 PMCID: PMC10274710 DOI: 10.1101/2023.06.03.543579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Type I interferons (IFNs) increase the excitability of dorsal root ganglion (DRG) neurons via activation of MNK-eIF4E translation signaling to promote pain sensitization in mice. Activation of STING signaling is a key component of type I IFN induction. Manipulation of STING signaling is an active area of investigation in cancer and other therapeutic areas. Vinorelbine is a chemotherapeutic that activates STING and has been shown to cause pain and neuropathy in oncology clinical trials in patients. There are conflicting reports on whether STING signaling promotes or inhibits pain in mice. We hypothesized that vinorelbine would cause a neuropathic pain-like state in mice via STING and signaling pathways in DRG neurons associated with type I IFN induction. Vinorelbine (10 mg/kg, i.v.) induced tactile allodynia and grimacing in WT male and female mice and increased p-IRF3 and type I IFN protein in peripheral nerves. In support of our hypothesis, vinorelbine-mediated pain was absent in male and female StingGt/Gt mice. Vinorelbine also failed to induce IRF3 and type I IFN signaling in these mice. Since type I IFNs engage translational control via MNK1-eIF4E in DRG nociceptors, we assessed vinorelbine-mediated p-eIF4E changes. Vinorelbine increased p-eIF4E in DRG in WT animals but not in StingGt/Gt or Mknk1-/- (MNK1 KO) mice. Consistent with these biochemical findings, vinorelbine had an attenuated pro-nociceptive effect in male and female MNK1 KO mice. Our findings support the conclusion that activation of STING signaling in the peripheral nervous system causes a neuropathic pain-like state that is mediated by type I IFN signaling to DRG nociceptors.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Eric T. David
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Krish J. Patel
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Abhira Ravirala
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
5
|
Tan PH, Ji J, Hsing CH, Tan R, Ji RR. Emerging Roles of Type-I Interferons in Neuroinflammation, Neurological Diseases, and Long-Haul COVID. Int J Mol Sci 2022; 23:ijms232214394. [PMID: 36430870 PMCID: PMC9696119 DOI: 10.3390/ijms232214394] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Interferons (IFNs) are pleiotropic cytokines originally identified for their antiviral activity. IFN-α and IFN-β are both type I IFNs that have been used to treat neurological diseases such as multiple sclerosis. Microglia, astrocytes, as well as neurons in the central and peripheral nervous systems, including spinal cord neurons and dorsal root ganglion neurons, express type I IFN receptors (IFNARs). Type I IFNs play an active role in regulating cognition, aging, depression, and neurodegenerative diseases. Notably, by suppressing neuronal activity and synaptic transmission, IFN-α and IFN-β produced potent analgesia. In this article, we discuss the role of type I IFNs in cognition, neurodegenerative diseases, and pain with a focus on neuroinflammation and neuro-glial interactions and their effects on cognition, neurodegenerative diseases, and pain. The role of type I IFNs in long-haul COVID-associated neurological disorders is also discussed. Insights into type I IFN signaling in neurons and non-neuronal cells will improve our treatments of neurological disorders in various disease conditions.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, MA 02482, USA
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Radika Tan
- Kaohsiung American School, Kaohsiung 81354, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
6
|
Abstract
Neuropsychiatric diseases have traditionally been studied from brain, and mind-centric perspectives. However, mounting epidemiological and clinical evidence shows a strong correlation of neuropsychiatric manifestations with immune system activation, suggesting a likely mechanistic interaction between the immune and nervous systems in mediating neuropsychiatric disease. Indeed, immune mediators such as cytokines, antibodies, and complement proteins have been shown to affect various cellular members of the central nervous system in multitudinous ways, such as by modulating neuronal firing rates, inducing cellular apoptosis, or triggering synaptic pruning. These observations have in turn led to the exciting development of clinical therapies aiming to harness this neuro-immune interaction for the treatment of neuropsychiatric disease and symptoms. Besides the clinic, important theoretical fundamentals can be drawn from the immune system and applied to our understanding of the brain and neuropsychiatric disease. These new frameworks could lead to novel insights in the field and further potentiate the development of future therapies to treat neuropsychiatric disease.
Collapse
|
7
|
Tan PH, Ji J, Yeh CC, Ji RR. Interferons in Pain and Infections: Emerging Roles in Neuro-Immune and Neuro-Glial Interactions. Front Immunol 2021; 12:783725. [PMID: 34804074 PMCID: PMC8602180 DOI: 10.3389/fimmu.2021.783725] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Interferons (IFNs) are cytokines that possess antiviral, antiproliferative, and immunomodulatory actions. IFN-α and IFN-β are two major family members of type-I IFNs and are used to treat diseases, including hepatitis and multiple sclerosis. Emerging evidence suggests that type-I IFN receptors (IFNARs) are also expressed by microglia, astrocytes, and neurons in the central and peripheral nervous systems. Apart from canonical transcriptional regulations, IFN-α and IFN-β can rapidly suppress neuronal activity and synaptic transmission via non-genomic regulation, leading to potent analgesia. IFN-γ is the only member of the type-II IFN family and induces central sensitization and microglia activation in persistent pain. We discuss how type-I and type-II IFNs regulate pain and infection via neuro-immune modulations, with special focus on neuroinflammation and neuro-glial interactions. We also highlight distinct roles of type-I IFNs in the peripheral and central nervous system. Insights into IFN signaling in nociceptors and their distinct actions in physiological vs. pathological and acute vs. chronic conditions will improve our treatments of pain after surgeries, traumas, and infections.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts, MA, United States
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Chun-Chang Yeh
- Department of Anesthesiology of Tri-Service General Hospital & National Defense Medical Center, Taipei City, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
- Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
8
|
Liu CC, Lu IC, Wang LK, Chen JY, Li YY, Yang CP, Liu PH, Cheng WJ, Tan PH. Interferon-β suppresses inflammatory pain through activating µ-opioid receptor. Mol Pain 2021; 17:17448069211045211. [PMID: 34517736 PMCID: PMC8642049 DOI: 10.1177/17448069211045211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Interferons (IFNs) are cytokines secreted by infected cells that can interfere with viral replication. Besides activating antiviral defenses, type I IFNs also exhibit diverse biological functions. IFN-β has been shown to have a protective effect against neurotoxic and inflammatory insults on neurons. Therefore, we aimed to investigate the possible role of IFN-β in reducing mechanical allodynia caused by Complete Freund's Adjuvant (CFA) injection in rats. We assessed the antinociceptive effect of intrathecal IFN-β in naïve rats and the rats with CFA-induced inflammatory pain. After the behavioral test, the spinal cords of the rats were harvested for western blot and immunohistochemical double staining. We found that intrathecal administration of IFN-β in naïve rats can significantly increase the paw withdrawal threshold and paw withdrawal latency. Further, the intrathecal injection of a neutralizing IFN-β antibody can reduce the paw withdrawal threshold and paw withdrawal latency, suggesting that IFN-β is produced in the spinal cord in normal conditions and serves as a tonic inhibitor of pain. In addition, intrathecal injection of IFN-β at dosages from 1000 U to 10000 U demonstrates a significant transient dose-dependent inhibition of CFA-induced inflammatory pain. This analgesic effect is reversed by intrathecal naloxone, suggesting that IFN-β produces an analgesic effect through central opioid receptor-mediated signaling. Increased expression of phospho-µ-opioid receptors after IFN-β injection was observed on western blot, and immunohistochemical staining showed that µ-opioids co-localized with IFN-α/βR in the dorsal horn of the spinal cord. The findings of this study demonstrate that the analgesic effect of IFN-β is through µ-opioid receptors activation in spial cord.
Collapse
Affiliation(s)
- Chien Cheng Liu
- Department of Anesthesiology, E-Da Hospital/I-Shou University, Kaohsiung City, Taiwan
| | - I Cheng Lu
- Department of Anesthesiology, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Li Kai Wang
- Southern Taiwan University of Science and Technology, Tainan City, Taiwan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Jen Yin Chen
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Yu Yu Li
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Chih Ping Yang
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ping Hsin Liu
- Department of Anesthesiology, E-Da Hospital/I-Shou University, Kaohsiung City, Taiwan
| | - Wan Jung Cheng
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ping Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| |
Collapse
|
9
|
Aw E, Zhang Y, Carroll M. Microglial responses to peripheral type 1 interferon. J Neuroinflammation 2020; 17:340. [PMID: 33183319 PMCID: PMC7659169 DOI: 10.1186/s12974-020-02003-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Interferon α (IFNα) is a cytokine whose production is increased endogenously in response to viral infection and in autoimmune diseases such as systemic lupus erythematosus (SLE). An elevated IFNα signature has been associated with clinically observed neuro-behavioural deficits such as mild cognitive impairment, fatigue, depression and psychosis in these diseases. However, the mechanisms underlying these neuropsychiatric symptoms remain largely unknown, and it is as yet unclear how IFNα signalling might influence central nervous system (CNS) function. Aberrant microglia-mediated synaptic pruning and function has recently been implicated in several neurodegenerative and neuropsychiatric diseases, but whether and how IFNα modulates these functions are not well defined. METHODS Using a model of peripheral IFNα administration, we investigated gene expression changes due to IFNAR signalling in microglia. Bulk RNA sequencing on sorted microglia from wild type and microglia-specific Ifnar1 conditional knockout mice was performed to evaluate IFNα and IFNAR signalling-dependent changes in gene expression. Furthermore, the effects of IFNα on microglia morphology and synapse engulfment were assessed, via immunohistochemistry and flow cytometry. RESULTS We found that IFNα exposure through the periphery induces a unique gene signature in microglia that includes the expected upregulation of multiple interferon-stimulated genes (ISGs), as well as the complement component C4b. We additionally characterized several IFNα-dependent changes in microglial phenotype, including expression of CD45 and CD68, cellular morphology and presynaptic engulfment, that reveal subtle brain region-specific differences. Finally, by specifically knocking down expression of IFNAR1 on microglia, we show that these changes are largely attributable to direct IFNAR signalling on microglia and not from indirect signalling effects through other CNS parenchymal cell types which are capable of IFNα-IFNAR signal transduction. CONCLUSIONS Peripheral IFNα induces unique genetic and phenotypic changes in microglia that are largely dependent on direct signalling through microglial IFNAR. The IFNα-induced upregulation of C4b could play important roles in the context of aberrant synaptic pruning in neuropsychiatric disease.
Collapse
Affiliation(s)
- Ernest Aw
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Yingying Zhang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Michael Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
10
|
Herzberg D, Strobel P, Ramirez-Reveco A, Werner M, Bustamante H. Chronic Inflammatory Lameness Increases Cytokine Concentration in the Spinal Cord of Dairy Cows. Front Vet Sci 2020; 7:125. [PMID: 32185190 PMCID: PMC7058553 DOI: 10.3389/fvets.2020.00125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/19/2020] [Indexed: 12/20/2022] Open
Abstract
Lameness in dairy cows is an extremely painful multifactorial condition that affects the welfare of animals and economically impacts the dairy industry worldwide. The aim of this study was to determine the profile of cytokines in the spinal cord dorsal horn of dairy cows with painful chronic inflammatory lameness. Concentrations of 10 cytokines were measured in the spinal cord of seven adult dairy cows with chronic lameness and seven adult dairy cows with no lameness. In all cows lameness was evaluated using a mobility scoring system and registered accordingly. Immediately after euthanasia the spinal cord was removed and 20 cm of lumbar segments (L2–L5) were obtained. After dorsal horn removal and processing, cytokine quantification of tumor necrosis factor-alpha (TNF-α), interleukin-1alpha (IL-1α), interleukin 13 (IL-13), chemokine-10 (CXCL10/IP-10), chemokine-9 (CXCL9/MIG), interferon-alpha (IFN-α), interferon-gamma (IFN-γ), interleukin-21 (IL-21), interleukin-36ra (IL-36ra), and macrophage inflammatory protein-1 beta (MIP-1β) was performed using a multiplex array. Lame cows had higher concentrations of TNF-α, IL-1-α, IL-13, CXCL10, CXCL9, IFN-α, and IFN-γ in their dorsal horn compared to non-lame cows, while IL-21 concentration was decreased. No differences in IL-36ra and MIP-1β concentrations between lame and non-lame cows were observed. Painful chronic inflammation of the hoof in dairy cows leads to a marked increase in cytokine concentration in the dorsal horn of the spinal cord, which could represent a state of neuroinflammation of the Central Nervous System (CNS).
Collapse
Affiliation(s)
- Daniel Herzberg
- Faculty of Veterinary Sciences, Graduate School, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Strobel
- Faculty of Veterinary Sciences, Animal Science Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Alfredo Ramirez-Reveco
- Faculty of Veterinary Sciences, Animal Science Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Marianne Werner
- Faculty of Veterinary Sciences, Animal Science Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Hedie Bustamante
- Faculty of Veterinary Sciences, Veterinary Clinical Sciences Institute, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
11
|
Fitzgibbon M, Kerr DM, Henry RJ, Finn DP, Roche M. Endocannabinoid modulation of inflammatory hyperalgesia in the IFN-α mouse model of depression. Brain Behav Immun 2019; 82:372-381. [PMID: 31505257 DOI: 10.1016/j.bbi.2019.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
Depression is a well-recognised effect of long-term treatment with interferon-alpha (IFN-α), a widely used treatment for chronic viral hepatitis and malignancy. In addition to the emotional disturbances, high incidences of painful symptoms such as headache and joint pain have also been reported following IFN-α treatment. The endocannabinoid system plays an important role in emotional and nociceptive processing, however it is unknown whether repeated IFN-α administration induces alterations in this system. The present study investigated nociceptive responding in the IFN-α-induced mouse model of depression and associated changes in the endocannabinoid system. Furthermore, the effects of modulating peripheral endocannabinoid tone on inflammatory pain-related behaviour in the IFN-α model was examined. Repeated IFN-α administration (8000 IU/g/day) to male C57/Bl6 mice increased immobility in the forced swim test and reduced sucrose preference, without altering body weight gain or locomotor activity, confirming development of the depressive-like phenotype. There was no effect of repeated IFN-α administration on latency to respond in the hot plate test on day 4 or 7 of treatment, however, formalin-evoked nociceptive behaviour was significantly increased in IFN-α treated mice following 8 days of IFN-α administration. 2-Arachidonoyl glycerol (2-AG) levels in the periaqueductal grey (PAG) and rostroventromedial medulla (RVM), and anandamide (AEA) levels in the RVM, were significantly increased in IFN-α-, but not saline-, treated mice following formalin administration. There was no change in endocannabinoid levels in the prefrontal cortex, spinal cord or paw tissue between saline- or IFNα-treated mice in the presence or absence of formalin. Furthermore, repeated IFN-α and/or formalin administration did not alter mRNA expression of genes encoding the endocannabinoid catabolic enzymes (fatty acid amide hydrolase or monoacylglycerol lipase) or endocannabinoid receptor targets (CB1, CB2 or PPARs) in the brain, spinal cord or paw tissue. Intra plantar administration of PF3845 (1 μg/10 μl) or MJN110 (1 μg/10 μl), inhibitors of AEA and 2-AG catabolism respectively, attenuated formalin-evoked hyperalgesia in IFN-α, but not saline-, treated mice. In summary, increasing peripheral endocannabinoid tone attenuates inflammatory hyperalgesia induced following repeated IFN-α administration. These data provide support for the endocannabinoid system in mediating and modulating heightened pain responding associated with IFNα-induced depression.
Collapse
Affiliation(s)
- Marie Fitzgibbon
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Daniel M Kerr
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland; Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland
| | - Rebecca J Henry
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland; NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland; NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland.
| |
Collapse
|
12
|
Woller SA, Ocheltree C, Wong SY, Bui A, Fujita Y, Gonçalves Dos Santos G, Yaksh TL, Corr M. Neuraxial TNF and IFN-beta co-modulate persistent allodynia in arthritic mice. Brain Behav Immun 2019; 76:151-158. [PMID: 30465880 PMCID: PMC6396982 DOI: 10.1016/j.bbi.2018.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 11/03/2018] [Accepted: 11/18/2018] [Indexed: 02/06/2023] Open
Abstract
In rheumatoid arthritis, joint pain can persist despite resolution of swelling. Similarly, in the murine K/BxN serum transfer model, a persistent tactile allodynia is observed after the resolution of joint inflammation (post-inflammatory pain) in male mice. Here, we found female wild type (WT) mice show inflammatory, but reduced post-inflammatory tactile allodynia. The transition to the post-inflammatory phenotype is dependent on TLR4 signaling. At the spinal level, we found differences in TNF and IFNβ mRNA expression in WT and TLR4 deficient males. In wild type male and female mice, there is differential temporal spinal expression of TNF and IFNβ. In WT males, blockade of TNF or administration of IFNβ was insufficient to affect the persistent allodynia. However, co-administration of intrathecal (IT) IFNβ and anti-TNF antibodies in male WT mice permanently reversed tactile allodynia. IT IFNβ treatment induces expression of anti-inflammatory proteins, contributing to the beneficial effect. Together, these experiments illustrated differences in the transition to chronic tactile allodynia in male and female animals and the complexities of effective pharmacologic interventions.
Collapse
Affiliation(s)
- Sarah A Woller
- Departments of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Cody Ocheltree
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | - Stephanie Y Wong
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | - Anthony Bui
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | - Yuya Fujita
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | | | - Tony L Yaksh
- Departments of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
13
|
Antagonising TLR4-TRIF signalling before or after a low-dose alcohol binge during adolescence prevents alcohol drinking but not seeking behaviour in adulthood. Neuropharmacology 2017; 128:460-473. [PMID: 28947376 DOI: 10.1016/j.neuropharm.2017.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/18/2017] [Accepted: 09/19/2017] [Indexed: 12/20/2022]
Abstract
Adolescents frequently engage in risky behaviours such as binge drinking. Binge drinking, in turn, perturbs neurodevelopment reinforcing reward seeking behaviour in adulthood. Current animal models are limited in their portrayal of this behaviour and the assessment of neuroimmune involvement (specifically the role of Toll-like receptor 4 (TLR4)). Therefore, the aims of this project were to develop a more relevant animal model of adolescent alcohol exposure and to characterise its effects on TLR4 signalling and alcohol-related behaviours later life. Balb/c mice received a short (P22-P25), low dose alcohol binge during in early adolescence, and underwent tests to investigate anxiety (elevated plus maze), alcohol seeking (conditioned place preference) and binge drinking behaviour (drinking in the dark) in adulthood. Four doses of alcohol during adolescence increased alcohol-induced conditioned place preference and alcohol intake in adulthood. However, this model did not affect basal elevated plus maze performance. Subsequent analysis of nucleus accumbal mRNA, revealed increased expression of TLR4-related mRNAs in mice who received alcohol during adolescence. To further elucidate the role of TLR4, (+)-Naltrexone, a biased TLR4 antagonist was administered 30 min before or after the adolescent binge paradigm. When tested in adulthood, (+)-Naltrexone treated mice exhibited reduced alcohol intake however, alcohol seeking and anxiety behaviour was unaltered. This study highlights that even a small amount of alcohol, when given during a critical neurodevelopmental period, can potentiate alcohol-related behaviours and TLR4 activation later in life. Interestingly, attenuation of TLR4 before or after adolescent alcohol exposure reduced only binge alcohol intake in adulthood.
Collapse
|
14
|
Zhang KJ, Yin XF, Yang YQ, Li HL, Xu YN, Chen LY, Liu XJ, Yuan SJ, Fang XL, Xiao J, Wu S, Xu HN, Chu L, Katlinski KV, Katlinskaya YV, Guo RB, Wei GW, Wang DC, Liu XY, Fuchs SY. A Potent In Vivo Antitumor Efficacy of Novel Recombinant Type I Interferon. Clin Cancer Res 2017; 23:2038-2049. [PMID: 27683179 PMCID: PMC5373932 DOI: 10.1158/1078-0432.ccr-16-1386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/28/2016] [Accepted: 09/11/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Antiproliferative, antiviral, and immunomodulatory activities of endogenous type I IFNs (IFN1) prompt the design of recombinant IFN1 for therapeutic purposes. However, most of the designed IFNs exhibited suboptimal therapeutic efficacies against solid tumors. Here, we report evaluation of the in vitro and in vivo antitumorigenic activities of a novel recombinant IFN termed sIFN-I.Experimental Design: We compared primary and tertiary structures of sIFN-I with its parental human IFNα-2b, as well as affinities of these ligands for IFN1 receptor chains and pharmacokinetics. These IFN1 species were also compared for their ability to induce JAK-STAT signaling and expression of the IFN1-stimulated genes and to elicit antitumorigenic effects. Effects of sIFN-I on tumor angiogenesis and immune infiltration were also tested in transplanted and genetically engineered immunocompetent mouse models.Results: sIFN-I displayed greater affinity for IFNAR1 (over IFNAR2) chain of the IFN1 receptor and elicited a greater extent of IFN1 signaling and expression of IFN-inducible genes in human cells. Unlike IFNα-2b, sIFN-I induced JAK-STAT signaling in mouse cells and exhibited an extended half-life in mice. Treatment with sIFN-I inhibited intratumoral angiogenesis, increased CD8+ T-cell infiltration, and robustly suppressed growth of transplantable and genetically engineered tumors in immunodeficient and immunocompetent mice.Conclusions: These findings define sIFN-I as a novel recombinant IFN1 with potent preclinical antitumorigenic effects against solid tumor, thereby prompting the assessment of sIFN-I clinical efficacy in humans. Clin Cancer Res; 23(8); 2038-49. ©2016 AACR.
Collapse
Affiliation(s)
- Kang-Jian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, Sichuan, China
| | - Xiao-Fei Yin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan-Qin Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Hui-Ling Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan-Ni Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lie-Yang Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xi-Jun Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Su-Jing Yuan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xian-Long Fang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuai Wu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hai-Neng Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Liang Chu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | - Yuliya V Katlinskaya
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rong-Bing Guo
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, Sichuan, China
| | - Guang-Wen Wei
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, Sichuan, China
| | - Da-Cheng Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xin-Yuan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
15
|
Verburg-van Kemenade BML, Cohen N, Chadzinska M. Neuroendocrine-immune interaction: Evolutionarily conserved mechanisms that maintain allostasis in an ever-changing environment. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:2-23. [PMID: 27296493 DOI: 10.1016/j.dci.2016.05.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 05/02/2023]
Abstract
It has now become accepted that the immune system and neuroendocrine system form an integrated part of our physiology. Immunological defense mechanisms act in concert with physiological processes like growth and reproduction, energy intake and metabolism, as well as neuronal development. Not only are psychological and environmental stressors communicated to the immune system, but also, vice versa, the immune response and adaptation to a current pathogen challenge are communicated to the entire body, including the brain, to evoke adaptive responses (e.g., fever, sickness behavior) that ensure allocation of energy to fight the pathogen. This phenomenon is evolutionarily conserved. Hence it is both interesting and important to consider the evolutionary history of this bi-directional neuroendocrine-immune communication to reveal phylogenetically ancient or relatively recently acquired mechanisms. Indeed, such considerations have already disclosed an extensive "common vocabulary" of information pathways as well as molecules and their receptors used by both the neuroendocrine and immune systems. This review focuses on the principal mechanisms of bi-directional communication and the evidence for evolutionary conservation of the important physiological pathways involved.
Collapse
Affiliation(s)
- B M Lidy Verburg-van Kemenade
- Cell Biology and Immunology Group, Dept. of Animal Sciences, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
| | - Nicholas Cohen
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| |
Collapse
|
16
|
Weber KT, Satoh S, Alipui DO, Virojanapa J, Levine M, Sison C, Quraishi S, Bloom O, Chahine NO. Exploratory study for identifying systemic biomarkers that correlate with pain response in patients with intervertebral disc disorders. Immunol Res 2015; 63:170-80. [PMID: 26440592 PMCID: PMC4689741 DOI: 10.1007/s12026-015-8709-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Molecular events that drive disc damage and low back pain (LBP) may precede clinical manifestation of disease onset and can cause detrimental long-term effects such as disability. Biomarkers serve as objective molecular indicators of pathological processes. The goal of this study is to identify systemic biochemical factors as predictors of response to treatment of LBP with epidural steroid injection (ESI). Since inflammation plays a pivotal role in LBP, this pilot study investigates the effect of ESI on systemic levels of 48 inflammatory biochemical factors (cytokines, chemokines, and growth factors) and examines the relationship between biochemical factor levels and pain or disability in patients with disc herniation (DH), or other diagnoses (Other Dx) leading to low back pain, which included spinal stenosis (SS) and degenerative disc disease (DDD). Study participants (n = 16) were recruited from a back pain management practice. Pain numerical rating score (NRS), Oswestry Disability Index (ODI), and blood samples were collected pre- and at 7 to 10 days post-treatment. Blood samples were assayed for inflammatory mediators using commercial multiplex assays. Mediator levels were compared pre- and post-treatment to investigate the potential correlations between clinical and biochemical outcomes. Our results indicate that a single ESI significantly decreased systemic levels of SCGF-β and IL-2. Improvement in pain in all subjects was correlated with changes in chemokines (MCP-1, MIG), hematopoietic progenitor factors (SCGF-β), and factors that participate in angiogenesis/fibrosis (HGF), nociception (SCF, IFN-α2), and inflammation (IL-6, IL-10, IL-18, TRAIL). Levels of biochemical mediators varied based on diagnosis of LBP, and changes in pain responses and systemic mediators from pre- to post-treatment were dependent on the diagnosis cohort. In the DH cohort, levels of IL-17 and VEGF significantly decreased post-treatment. In the Other Dx cohort, levels of IL-2Rα, IL-3, and SCGF-β significantly decreased post-treatment. In order to determine whether mediator changes were related to pain, correlations between change in pain scores and change in mediator levels were performed. Subjects with DH demonstrated a profile signature that implicated hematopoiesis factors (SCGF-β, GM-CSF) in pain response, while subjects with Other Dx demonstrated a biomarker profile that implicated chemokines (MCP-1, MIG) and angiogenic factors (HGF, VEGF) in pain response. Our findings provide evidence that systemic biochemical factors in patients with LBP vary by diagnosis, and pain response to treatment is associated with a unique profile of biochemical responses in each diagnosis group. Future hypothesis-based studies with larger subject cohorts are warranted to confirm the findings of this pilot exploratory study.
Collapse
Affiliation(s)
- K T Weber
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shina Satoh
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - D Olivier Alipui
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Justin Virojanapa
- Department of Neurosurgery, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
| | - Mitchell Levine
- Department of Neurosurgery, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
| | - Cristina Sison
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Molecular Medicine, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
| | - Shaheda Quraishi
- Department of Neurosurgery, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
- Department of Physical Medicine and Rehabilitation, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
| | - Ona Bloom
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Physical Medicine and Rehabilitation, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
- Department of Molecular Medicine, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA
| | - Nadeen O Chahine
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Department of Neurosurgery, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA.
- Department of Molecular Medicine, Hofstra North Shore LIJ School of Medicine, Hempstead, NY, USA.
| |
Collapse
|
17
|
Mechanisms for interferon-α-induced depression and neural stem cell dysfunction. Stem Cell Reports 2014; 3:73-84. [PMID: 25068123 PMCID: PMC4110771 DOI: 10.1016/j.stemcr.2014.05.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 11/23/2022] Open
Abstract
New neurons generated by the neural stem cells (NSCs) in the adult hippocampus play an important role in emotional regulation and respond to the action of antidepressants. Depression is a common and serious side effect of interferon-α (IFN-α), which limits its use as an antiviral and antitumor drug. However, the mechanism(s) underlying IFN-induced depression are largely unknown. Using a comprehensive battery of behavioral tests, we found that mice subjected to IFN-α treatment exhibited a depression-like phenotype. IFN-α directly suppressed NSC proliferation, resulting in the reduced generation of new neurons. Brain-specific mouse knockout of the IFN-α receptor prevented IFN-α-induced depressive behavioral phenotypes and the inhibition of neurogenesis, suggesting that IFN-α suppresses hippocampal neurogenesis and induces depression via its receptor in the brain. These findings provide insight for understanding the neuropathology underlying IFN-α-induced depression and for developing new strategies for the prevention and treatment of IFN-α-induced depressive effects. IFN-α-treated mice show a depression-like phenotype in a behavioral test battery IFN-α directly suppresses NSC proliferation in adult hippocampus IFN-α suppresses neurogenesis and induced depression via its receptor in the brain
Collapse
|
18
|
Tan PH, Gao YJ, Berta T, Xu ZZ, Ji RR. Short small-interfering RNAs produce interferon-α-mediated analgesia. Br J Anaesth 2012; 108:662-9. [PMID: 22307241 DOI: 10.1093/bja/aer492] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND There is increasing interest in RNA interference in pain research using the intrathecal route to deliver small-interfering RNA (siRNA). An interferon (IFN) response is a common side-effect of siRNA. However, the IFN response in the spinal cord after intrathecal administration of siRNA remains unknown. We hypothesized that high doses of siRNAs can elicit off-target analgesia via releasing IFN-α. We investigated the IFN response and its role in regulating pain sensitivity in the spinal cords after intrathecal administration of siRNAs. METHODS Male Sprague-Dawley rats were given intrathecal injections of non-targeting (NT) siRNAs or IFN-α and tested for complete Freund's adjuvant (CFA)-induced mechanical allodynia and heat hyperalgesia. IFN-α in the spinal cord after injection of NT siRNAs was measured by western blotting and immunohistochemical staining. RESULTS IFN-α was up-regulated in the spinal cord after intrathecal treatment of NT siRNAs. Intrathecal injection of NT siRNAs, at high doses of 10 or 20 μg, reduced CFA-induced inflammatory pain (P<0.05). Intrathecal application of IFN-α inhibited pain hypersensitivity in inflamed rats and produced analgesia in naïve rats (P<0.05). Notably, the anti-nociceptive effects elicited by NT siRNAs and IFN-α were reversed by IFN-α neutralizing antibody and naloxone. CONCLUSIONS Our data suggest that (i) intrathecal administration of high doses of siRNA (≥ 10 μg) induced up-regulation of IFN-α in the spinal cord and produced analgesic effects through IFN-α, and (ii) IFN-α's analgesic effect is mediated via opioid receptors. Caution must be taken to avoid IFN-α-mediated analgesic effects of siRNAs in pain research.
Collapse
Affiliation(s)
- P H Tan
- Department of Anesthesiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
19
|
Qin ZF, Hou DY, Fang YQ, Xiao HJ, Wang J, Li KC. Interferon-alpha enhances excitatory transmission in substantia gelatinosa neurons of rat spinal cord. Neuroimmunomodulation 2012; 19:235-40. [PMID: 22441540 DOI: 10.1159/000335167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 11/16/2011] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE It has been shown that interferon-α (IFN-α) is synthesized and secreted by macrophages, monocytes, T lymphocytes, glial cells and neurons. IFN-α has been shown to have an antinociceptive effect at the supraspinal level in the nerve system. However, it is unclear how IFN-α is involved in the modulation of nociceptive transmission in the spinal cord. METHODS In the present study, IFN-α was used to test the potential functional roles in the nociceptive transmission. Using the whole-cell patch-clamp technique, we examined the effects of IFN-α on substantia gelatinosa (SG) neurons in the dorsal root-attached spinal cord slice prepared from adult rats. RESULTS We found that IFN-α increased glutamatergic excitatory postsynaptic currents evoked by the stimulation of either Aδ or C afferent fibers. Further studies showed that IFN-α treatment dose-dependently increased spontaneous excitatory postsynaptic current frequency in SG neurons, while not affecting the amplitude. Moreover, intrathecal antibody of IFN-α could reduce nociceptive responses in formalin test. CONCLUSIONS These results suggest that IFN-α presynaptically facilitates the excitatory synaptic transmission to SG neurons. The nociceptive responses could be inhibited by IFN-α antibody in the formalin test. Thus, IFN-α enhances the nociceptive transmission, which contributes to the behavioral nociceptive responses.
Collapse
Affiliation(s)
- Zhi-Feng Qin
- School of Aeronautic Science and Engineering, Beihang University, Beijing, China
| | | | | | | | | | | |
Collapse
|
20
|
Reyes-Vázquez C, Prieto-Gómez B, Dafny N. Interferon modulates central nervous system function. Brain Res 2011; 1442:76-89. [PMID: 22322149 DOI: 10.1016/j.brainres.2011.09.061] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/26/2011] [Accepted: 09/27/2011] [Indexed: 12/15/2022]
Abstract
The interferons (IFNs) are an endogenous pleiotropic family of cytokines that perform fundamental physiological functions as well as protecting host organisms from disease and in maintaining homeostasis. This review covers the effects of endogenous IFN on the nervous system. It starts with the description of its receptors, followed how it modulate neuronal activity, mood, sleep, temperature, the endocrine system, the opioid system and how it regulate food consumption and the immune system. Similar to other multifunctional cytokines, an excessive or inappropriate activity of IFNs can cause toxicity and even death. Furthermore, IFNs are currently the major treatment modality for several malignant and non-malignant diseases such as chronic hepatitis C and B, multiple sclerosis, hematological malignancies, malignant melanoma, renal cell carcinoma, etc.
Collapse
Affiliation(s)
- Cruz Reyes-Vázquez
- Departamento de Fisiología, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México Apdo. Postal 70250, México, 20, DF México
| | | | | |
Collapse
|
21
|
Lee JK, Park SH, Sim YB, Jung JS, Suh HW. Interaction of supraspinally administered interferon-alpha with opioid system in the production of antinociception. Arch Pharm Res 2010; 33:1059-63. [DOI: 10.1007/s12272-010-0712-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 04/08/2010] [Accepted: 04/12/2010] [Indexed: 10/19/2022]
|
22
|
Lang JY, Wang JS, Zhai HF, Fang Q, Wu P, Lu L. Interferon-alpha reinstates morphine-conditioned place preference through opioid receptors in rats. Behav Pharmacol 2009; 20:166-73. [DOI: 10.1097/fbp.0b013e32832a805e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Citak EC, Oguz A, Karadeniz C, Okur A, Memis L, Boyunaga O. Management of plexiform neurofibroma with interferon alpha. Pediatr Hematol Oncol 2008; 25:673-8. [PMID: 18850480 DOI: 10.1080/08880010802315983] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Plexiform neurofibroma is a relatively common but potentially devastating manifestation of neurofibromatosis type 1 (NF 1). A substantial number of plexiform neurofibroma causes morbidity. Various treatment modalities are considered to decrease pain. In this paper a case with plexiform neurofibroma causing severe pain and in whom alpha-interferon was used is presented.
Collapse
Affiliation(s)
- Elvan Caglar Citak
- Gazi University Faculty of Medicine, Department of Pediatric Oncology, Ankara, Turkey.
| | | | | | | | | | | |
Collapse
|
24
|
Wang JY, Zeng XY, Fan GX, Yuan YK, Tang JS. μ- but not δ- and κ-opioid receptor mediates the nucleus submedius interferon-α-evoked antinociception in the rat. Neurosci Lett 2006; 397:254-8. [PMID: 16406668 DOI: 10.1016/j.neulet.2005.12.046] [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: 11/08/2005] [Revised: 12/12/2005] [Accepted: 12/12/2005] [Indexed: 11/22/2022]
Abstract
Previous studies have indicated that interferon-alpha (IFN-alpha) can bind to opioid receptors and exerts an antinociceptive effect in both peripheral and central nervous systems. The current study investigated the antinociceptive effect of IFN-alpha unilaterally microinjected into the thalamic nucleus submedius (Sm) of rats on noxious thermal stimulus, and the roles of different subtypes of opioid receptors in mediating the Sm IFN-alpha-evoked antinociception. The results indicated that unilateral microinjection of IFN-alpha (4, 8, 16 pmol) into the Sm dose-dependently increased the hind paw withdrawal latency from the noxious heat stimulus, and this effect was reversed by pretreatment with non-selective opioid receptor antagonist naloxone (200 pmol) and specific mu-opioid receptor antagonist beta-FNA (1 nmol) into the same sites, whereas delta-opioid receptor antagonist ICI174,864 (1 nmol) and kappa-opioid receptor antagonist nor-BNI (1 nmol) failed to alter the effect of IFN-alpha. These results suggest that Sm is involved in IFN-alpha-evoked antinociception and mu- but not delta- and kappa-opioid receptor mediates the Sm IFN-alpha-evoked antinociception.
Collapse
Affiliation(s)
- Jun-Yang Wang
- Department of Immunology and Pathogenic Biology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, PR China
| | | | | | | | | |
Collapse
|
25
|
Dafny N, Yang PB. Interferon and the central nervous system. Eur J Pharmacol 2005; 523:1-15. [PMID: 16226745 DOI: 10.1016/j.ejphar.2005.08.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Revised: 07/01/2005] [Accepted: 08/18/2005] [Indexed: 01/14/2023]
Abstract
Interferons (IFNs) were discovered as natural antiviral substances produced during viral infection and were initially characterized for their ability to "interfere" with viral replication, slow cell proliferation, and profound alteration of immunity. The IFNs are synthesized and secreted by monocytes, macrophages, T-lymphocytes, neurons, and glia cells. The different IFNs are classified into three classes: alpha, beta, and gamma. alpha-IFN produced in the brain exerts direct effects on the brain and endocrine system by activating the neurosecretory hypothalamic neurons and regulates the hypothalamic-pituitary-adrenocortical axis. IFNs modulate neurophysiological activities of many brain region involving in pain, temperature, and food intake regulation. alpha-IFN administration activates the sympathetic nerves innervating components of the immune system. IFNs may serve as regulatory mediators between the central nervous system, the immune system, and endocrine system. IFN is used as immunologic therapy to treat various hematologic malignancies and infectious ailments and autoimmune diseases.
Collapse
Affiliation(s)
- Nachum Dafny
- Department of Neurobiology and Anatomy, The University of Texas, Medical School, P.O. Box 20708, Houston, Texas 77225, USA.
| | | |
Collapse
|
26
|
Abstract
One of the truly remarkable discoveries in modern biology is the finding that the nervous system and immune system use a common chemical language for intra- and inter-system communication. This review will discuss some of the pivotal results that deciphered this chemical language. Specifically the nervous and immune systems produce a common set of peptide and nonpeptide neurotransmitters and cytokines that act on a common repertoire of receptors in the two systems. The paper will also review more recent studies that have delineated hardwired and humoral pathways for such bidirectional communication. This is discussed in the context of the idea that the sharing of ligands and receptors allows the immune system to serve as the sixth sense that notifies the nervous system of the presence of entities, such as viruses and bacteria, that are imperceptible to the classic senses. Lastly, this review will suggest ways to apply the newfound knowledge of the sixth sense to understand a placebo effect and to treat human disease.
Collapse
Affiliation(s)
- J E Blalock
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
27
|
Kobayashi T, Washiyama K, Ikeda K. Effects of interferon-α on cloned opioid receptors expressed in Xenopus oocytes. Life Sci 2004; 76:407-15. [PMID: 15530503 DOI: 10.1016/j.lfs.2004.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Accepted: 07/17/2004] [Indexed: 11/30/2022]
Abstract
Interferon-alpha (IFNalpha) affects the opioid system. However, the direct action of IFNalpha on cloned opioid receptors remains unknown. Taking advantage of the functional coupling of cloned opioid receptors to G protein-activated inwardly rectifying K+ (GIRK) channels in a Xenopus oocyte expression system, we investigated the effects of recombinant IFNalpha on cloned mu-, delta- and kappa-opioid receptors. In oocytes co-injected with mRNAs for either the delta- or kappa-opioid receptor and for GIRK channel subunits, IFNalpha at high concentrations induced small GIRK currents that were abolished by naloxone, an opioid-receptor antagonist, compared with the control responses to each selective opioid agonist. Additionally, IFNalpha induced no significant current response in oocytes injected with mRNA(s) for either opioid receptor alone or GIRK channels. In oocytes expressing the mu-opioid receptor and GIRK channels, IFNalpha had little or no effect. Moreover, in oocytes expressing each opioid receptor and GIRK channels, GIRK current responses to each selective opioid agonist were not affected by the presence of IFNalpha, indicating no significant antagonism of IFNalpha toward the opioid receptors. Furthermore, IFNalpha had little or no effect on the mu/delta-, delta/kappa- or mu/kappa-opioid receptors expressed together with GIRK channels in oocytes. Our results suggest that IFNalpha weakly activates the delta and kappa-opioid receptors. The direct activation of the delta- and kappa-opioid receptors by IFNalpha may partly contribute to some of the IFNalpha effects under its high-dose medication.
Collapse
MESH Headings
- Animals
- Cloning, Molecular
- Dose-Response Relationship, Drug
- Female
- G Protein-Coupled Inwardly-Rectifying Potassium Channels
- Interferon-alpha/pharmacology
- Oocytes/drug effects
- Oocytes/metabolism
- Patch-Clamp Techniques
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/metabolism
- RNA, Messenger/metabolism
- Receptors, Opioid/drug effects
- Receptors, Opioid/genetics
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Recombinant Proteins
- Xenopus laevis
Collapse
Affiliation(s)
- Toru Kobayashi
- Department of Molecular Neuropathology, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, Niigata 951-8585, Japan.
| | | | | |
Collapse
|
28
|
Wang YX, Xu WG, Sun XJ, Chen YZ, Liu XY, Tang H, Jiang CL. Fever of recombinant human interferon-alpha is mediated by opioid domain interaction with opioid receptor inducing prostaglandin E2. J Neuroimmunol 2004; 156:107-12. [PMID: 15465601 DOI: 10.1016/j.jneuroim.2004.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 07/22/2004] [Accepted: 07/23/2004] [Indexed: 11/20/2022]
Abstract
We have reported that there are distinct domains in Interferon-alpha (IFNalpha) molecule mediating immune and opioid-like effects respectively. And the opioid effect of IFNalpha is mediated by mu opioid receptor. We report here the structural basis of fever induced by recombinant human IFNalpha. Two kinds of IFNalpha mutants were obtained and used to investigate the structural basis of fever of IFNalpha, which are 129Ser-IFNalpha and 38Leu-IFNalpha. The antiviral activity of 129Ser-IFNalpha almost disappeared, but there still retained the strong analgesic activity. The antiviral activity of 38Leu-IFNalpha remained, but the analgesic activity disappeared completely. It showed that IFNalpha and 129Ser-IFNalpha decreased cAMP production, induced the fever, and stimulated PGE2 to release from the hypothalamus slices, which could be blocked by naloxone, but 38Leu-IFNalpha failed. It is the first demonstration that fever induced by IFNalpha is mediated by opioid domain of IFNalpha interacting with opioid receptor. It is inferred that high-activity and low side-effect IFNalpha or other cytokines could be obtained after being changed the motifs in the tertiary structure.
Collapse
Affiliation(s)
- Yun-Xia Wang
- Department of Nautical Medicine, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, P.R. China
| | | | | | | | | | | | | |
Collapse
|
29
|
Cao CM, Xia Q, Tu J, Chen M, Wu S, Wong TM. Cardioprotection of Interleukin-2 Is Mediated via κ-Opioid Receptors. J Pharmacol Exp Ther 2004; 309:560-7. [PMID: 14747612 DOI: 10.1124/jpet.103.061135] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We examined whether interleukin-2 (IL-2) protects the myocardium against injury induced by ischemia and reperfusion via the kappa-opioid receptor (OR). The cardioprotective effect of IL-2 was evaluated by measuring infarct size and lactate dehydrogenase (LDH) release in response to ischemia and reperfusion in the isolated rat heart. IL-2 at an optimal dose of 50 U/ml mimicked the effect of ischemic preconditioning by reducing infarct size and LDH release. The infarct and LDH-reducing effects of IL-2 were blocked by nor-binaltorphimine (5 microM), a kappa-OR antagonist, but not naltrindole (5 microM), a delta-OR antagonist known to block the action of its stimulation. Moreover, blockade of the mitochondrial ATP-sensitive potassium (mito-K(ATP)) channel with a selective antagonist, 5-hydroxydecanoate (100 microM), or a nonselective antagonist of K(ATP) channels, glybenclamide (100 microM), or blockade of protein kinase C (PKC) with its inhibitors chelerythrine (5 microM) or GF 109203X (10 microM) [3-[1-[3-(dimethylaminopropyl]-1H-indol-3-yl]-4-(1H-indol-3-yl)-1H-pyrrole-2,5-dione monohydrochloride] abolished the protective effect of IL-2. Administration of free radical scavengers N-acetylcysteine (4 mM) or N-(2-mercaptopropionyl)-glycine (1 mM) also abolished the protective effects of IL-2 and U50,488H [(trans)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]benzeneacetamide], a selective kappa-OR agonist. This study provides the first evidence that IL-2 confers cardioprotection against injury induced by ischemia/reperfusion. The effect of IL-2 is mediated via kappa-OR as evidenced by kappa-OR antagonism and similar signaling mechanisms, mito-K(ATP), PKC, and reactive oxygen species involved in the cardioprotective effects of both IL-2 and kappa-OR stimulation.
Collapse
Affiliation(s)
- Chun-Mei Cao
- Department of Physiology, The University of Hong Kong, Hong Kong Special Administrative Region, SAR, China
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
This paper is the twenty-third installment of the annual review of research concerning the opiate system. It summarizes papers published during 2000 that studied the behavioral effects of the opiate peptides and antagonists, excluding the purely analgesic effects, although stress-induced analgesia is included. The specific topics covered this year include stress; tolerance and dependence; learning, memory, and reward; eating and drinking; alcohol and other drugs of abuse; sexual activity, pregnancy, and development; mental illness and mood; seizures and other neurological disorders; electrical-related activity; general activity and locomotion; gastrointestinal, renal, and hepatic function; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- A L Vaccarino
- Department of Psychology, University of New Orleans, New Orleans, LA 70148, USA.
| | | |
Collapse
|
31
|
You ZD, Li JH, Song CY, Lu CL, He C. Oxytocin mediates the inhibitory action of acute lithium on the morphine dependence in rats. Neurosci Res 2001; 41:143-50. [PMID: 11591442 DOI: 10.1016/s0168-0102(01)00272-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The role of central oxytocin in inhibitory action of lithium on the development of morphine dependence was behavioral investigated in rats. Acute lithium could enhance the morphine-induced analgesia in rats with or without chronic morphine treatment; this effect could be inhibited by intraventricular injection of oxytocin antagonist d (CH(2))(5)-Tyr (Me)-[Orn(8)]-Vasotocin (OVT). Lithium could attenuate naloxone-precipitated withdrawal signs in morphine dependent rats. The reduction of the expression of naloxone-precipitated withdrawal signs by lithium was reversed by ICV of OVT. The lithium significantly inhibited the conditioned place preference (CPP) induced by morphine, which inhibitory action of lithium could also reverse by ICV injection of OVT. These results suggested that lithium might inhibit the physical dependence on morphine as well as psychological dependence in rats, and that this inhibitory effect of lithium on the development of morphine dependence might be associated with oxytocin systems in the central nervous system.
Collapse
Affiliation(s)
- Z D You
- Department of Neurobiology, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, People's Republic of China.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Interferon-alpha (IFNalpha) is not only an immunoregulatory factor, but is also an analgesic molecule. We ever reported that there exist distinct domains in IFNalpha molecule that mediate immune and analgesic effects respectively and inferred that the analgesic domain locates around the 122nd Tyr residue of IFNalpha molecule in the tertiary structure. After the 36th Phe residue, which was located closely to the 122nd Tyr residue in the tertiary structure, was mutated to Ser using site-directed mutagenesis, the analgesic activity of this mutant lost completely, but the antiviral activity of IFNalpha still maintained 40.5% of wild type IFNalpha. The results suggest that the 36th Phe residue is one of the constituent for the analgesic domain of IFNalpha and inferred that the analgesic domain of IFNalpha consists of the 122nd Tyr and the residues around the 122nd in the tertiary structure, which include the 36th Phe.
Collapse
Affiliation(s)
- Y X Wang
- Department of Neurobiology, Institute of Neuroscience, China Medical University, Shenyang, PR China
| | | | | | | | | | | | | |
Collapse
|
33
|
Wang YX, Jiang CL, Lu CL, Song LX, You ZD, Shao XY, Cui RY, Liu XY. Distinct domains of IFNalpha mediate immune and analgesic effects respectively. J Neuroimmunol 2000; 108:64-7. [PMID: 10900338 DOI: 10.1016/s0165-5728(00)00271-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Interferon-alpha (IFNalpha) is not only an immunoregulatory factor, but is also an analgesic molecule. The analgesic effect of IFNalpha was mediated by mu opioid receptor. After the 129th Tyr residue of human IFNalpha was mutated to Ser, the antiviral activity almost disappeared, but there still remained a strong analgesic activity that could be blocked by naloxone. These results indicate that there exist distinct domains in the IFNalpha molecule, which mediate immune and analgesic effects respectively, and suggest that there are different receptor mechanisms inducing immune and analgesic effects of IFNalpha. However, although the antiviral activity of IFNalpha decreased to 34.1% of wild type IFNalpha after the 122nd Tyr residue was changed to Ser, the analgesic activity of this mutant was lost completely. There were significant cross reactivities between INFalpha and anti-opioid sera. These studies show strong structural and functional similarities between INFalpha and opioid peptides, and inferred that the analgesic domain locates around the 122nd Tyr residue of IFNalpha molecule in tertiary structure.
Collapse
Affiliation(s)
- Y X Wang
- Department of Neurobiology, Second Military Medical University, 800 Xiangyin Road, 200433, Shanghai, PR China
| | | | | | | | | | | | | | | |
Collapse
|