51
|
Reiss D, Maduna T, Maurin H, Audouard E, Gaveriaux-Ruff C. Mu opioid receptor in microglia contributes to morphine analgesic tolerance, hyperalgesia, and withdrawal in mice. J Neurosci Res 2020; 100:203-219. [PMID: 32253777 DOI: 10.1002/jnr.24626] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/23/2020] [Accepted: 03/18/2020] [Indexed: 12/17/2022]
Abstract
A major challenge in medicine is developing potent pain therapies without the adverse effects of opiates. Neuroinflammation and in particular microglial activation have been shown to contribute to these effects. However, the implication of the microglial mu opioid receptor (MOR) is not known. We developed a novel conditional knockout (cKO) mouse line, wherein MOR is deleted in microglia. Morphine analgesic tolerance was delayed in both sexes in cKO mice in the hot plate assay. Opioid-induced hyperalgesia (OIH) as measured in the tail immersion assay was abolished in male cKO mice, and physical dependence to morphine as assessed by naloxone-induced withdrawal was attenuated in female cKO mice. Our results show a sex-dependent contribution of microglial MOR in morphine analgesic tolerance, OIH, and physical dependence. In conclusion, our data suggest that blockade of microglial MOR could represent a therapeutic target for opiate analgesia without the opiate adverse effects.
Collapse
Affiliation(s)
- David Reiss
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Tando Maduna
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Neurology Research Group, Department of Physiology, Stellenbosch University, Stellenbosch, South Africa
| | - Hervé Maurin
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Emilie Audouard
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Claire Gaveriaux-Ruff
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
52
|
Uniyal A, Gadepalli A, Akhilesh, Tiwari V. Underpinning the Neurobiological Intricacies Associated with Opioid Tolerance. ACS Chem Neurosci 2020; 11:830-839. [PMID: 32083459 DOI: 10.1021/acschemneuro.0c00019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The opioid crisis is a major threat of the 21st century, with a remarkable juxtaposition of use and abuse. Opioids are the most potent and efficacious class of analgesics, but despite their proven therapeutic efficacy, they have recently been degraded to third-line therapy for the management of chronic pain in clinics. The reason behind this is the development of potential side effects and tolerance after repeated dosing. Opioid tolerance is the major limiting factor leading to the withdrawal of treatment, severe side effects due to dose escalation, and sometimes even death of the patients. Every day more than 90 people die due to opioids overdose in America, and a similar trend has been seen across the globe. Over the past two decades, researchers have been trying to dissect the neurobiological mechanism of opioid tolerance. Research on opioid tolerance shifted toward central nervous system-based adaptations because tolerance is much more than just a cellular phenomenon. Thus, neurobiological adaptations associated with opioid tolerance are important to understand in order to find newer pain therapeutics. These adaptations are associated with alterations in ascending and descending pain pathways, reward circuitry modulations, receptor desensitization and down-regulation, receptor internalization, heterodimerization, and altered epigenetic regulation. The present Review is focused on novel circuitries associated with opioid tolerance in different areas of the brain, such as periaqueductal gray, rostral ventromedial medulla, dorsal raphe nucleus, ventral tegmental area, and nucleus accumbens. Understanding the neurobiological modulations associated with chronic opioid exposure and tolerance will pave the way for the development of novel pharmacological tools for safer and better management of chronic pain in patients.
Collapse
Affiliation(s)
- Ankit Uniyal
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005, Uttar Pradesh, India
| | - Anagha Gadepalli
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005, Uttar Pradesh, India
| | - Akhilesh
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005, Uttar Pradesh, India
| |
Collapse
|
53
|
Dave RS, Ali H, Sil S, Knight LA, Pandey K, Madduri LSV, Qiu F, Ranga U, Buch S, Byrareddy SN. NF-κB Duplications in the Promoter-Variant HIV-1C LTR Impact Inflammation Without Altering Viral Replication in the Context of Simian Human Immunodeficiency Viruses and Opioid-Exposure. Front Immunol 2020; 11:95. [PMID: 32076422 PMCID: PMC7006833 DOI: 10.3389/fimmu.2020.00095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Recent spread of the promoter variant (4-κB) Human immunodeficiency virus-1 clade C (HIV-1C) strain is attributed to duplication of the Nuclear Factor Kappa B (NF-κB) binding sites and potential increased heroin consumption in India. To study the underlying biology of 4-κB HIV-1C in rhesus macaques, we engineered a promoter-chimera variant (4NF-κB) Simian Human Immunodeficiency Virus (SHIV) by substituting the HIV-1C Long Terminal Repeat (LTR) region consisting of 4 NF-κB and 3 Sp-1 sites with the corresponding segment in the LTR of SHIV AD8EO. The wild-type (3NF-κB) promoter-chimera SHIV was generated by inactivating the 5' proximal NF-κB binding site in SHIV 4NF-κB. CD8-depleted rhesus macaque PBMCs (RM-PBMCs) were infected with the promoter-chimera and AD8EO SHIVs to determine the effects of opioid-exposure on inflammation, NF-κB activation, neurotoxicity in neuronal cells and viral replication. Morphine-exposure of RM-PBMCs infected with SHIVs 4NF-κB, 3NF-κB, and AD8EO altered cellular transcript levels of monocyte chemoattractant protein 1, interleukin 6, interleukin 1β, and Tumor Necrosis Factor α. Of note, divergent alteration of the cytokine transcript levels was observed with these promoter-chimera wild-type and variant SHIVs. NF-κB activation was observed during infection of all three SHIVs with morphine-exposure. Finally, we observed that SHIV AD8EO infection and exposure to both morphine and naloxone had the greatest impact on the neurotoxicity. The promoter-chimera SHIV 4NF-κB and SHIV 3NF-κB did not have a similar effect on neurotoxicity as compared to SHIV AD8EO. All SHIVs replicated efficiently at comparable levels in RM-PBMCs and morphine-exposure did not alter viral replication kinetics. Future in vivo studies in rhesus macaques will provide greater understanding of 4-κB HIV-1C viral immunopathogenesis and onset of disease in the central nervous system during morphine-exposure.
Collapse
Affiliation(s)
- Rajnish S. Dave
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Haider Ali
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Center for Advanced Scientific Research, Bangalore, India
| | - Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lindsey A. Knight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lepakshe S. V. Madduri
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Fang Qiu
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Udaykumar Ranga
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Center for Advanced Scientific Research, Bangalore, India
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
54
|
Abstract
Research on the effects of opioids on immune responses was stimulated in the 1980s by the intersection of use of intravenous heroin and HIV infection, to determine if opioids were enhancing HIV progression. The majority of experiments administering opioid alkaloids (morphine and heroin) in vivo, or adding these drugs to cell cultures in vitro, showed that they were immunosuppressive. Immunosuppression was reported as down-regulation: of Natural Killer cell activity; of responses of T and B cells to mitogens; of antibody formation in vivo and in vitro; of depression of phagocytic and microbicidal activity of neutrophils and macrophages; of cytokine and chemokine production by macrophages, microglia, and astrocytes; by sensitization to various infections using animal models; and by enhanced replication of HIV in vitro. The specificity of the receptor involved in the immunosuppression was shown to be the mu opioid receptor (MOR) by using pharmacological antagonists and mice genetically deficient in MOR. Beginning with a paper published in 2005, evidence was presented that morphine is immune-stimulating via binding to MD2, a molecule associated with Toll-like Receptor 4 (TLR4), the receptor for bacterial lipopolysaccharide (LPS). This concept was pursued to implicate inflammation as a mechanism for the psychoactive effects of the opioid. This review considers the validity of this hypothesis and concludes that it is hard to sustain. The experiments demonstrating immunosuppression were carried out in vivo in rodent strains with normal levels of TLR4, or involved use of cells taken from animals that were wild-type for expression of TLR4. Since engagement of TLR4 is universally accepted to result in immune activation by up-regulation of NF-κB, if morphine were binding to TLR4, it would be predicted that opioids would have been found to be pro-inflammatory, which they were not. Further, morphine is immunosuppressive in mice with a defective TLR4 receptor. Morphine and morphine withdrawal have been shown to permit leakage of Gram-negative bacteria and LPS from the intestinal lumen. LPS is the major ligand for TLR4. It is proposed that an occult variable in experiments where morphine is being proposed to activate TLR4 is actually underlying sepsis induced by the opioid.
Collapse
Affiliation(s)
- Toby K. Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
55
|
Zhang L, Dong L, Tang Y, Li M, Zhang M. MiR-146b protects against the inflammation injury in pediatric pneumonia through MyD88/NF-κB signaling pathway. Infect Dis (Lond) 2019; 52:23-32. [PMID: 31583932 DOI: 10.1080/23744235.2019.1671987] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Pneumonia is a common respiratory disease worldwide that can be prevented and treated. However, it is considered to be the leading cause of children death. The present study was aimed to explore the functional role and molecular mechanism of miR-146b in the inflammation injury in pediatric pneumonia.Materials and methods: The lipopolysaccharide (LPS)-induced pulmonary injury cell model was established in WI-38 human lung fibroblasts cells. QRT-PCR and Western blot was applied to detect miR-146b and MyD88 expression. ELISA assay was used to analyze the production of pro-inflammatory factors. Cell viability was evaluated by CCK-8 assay. The apoptosis proteins and the downstream genes of NF-κB pathway were detected by Western blot.Results: we displayed that miR-146b was down-regulated, whereas MyD88 was up-regulated in the serum of children patients with pneumonia and in WI-38 cells treated with LPS. Moreover, re-expression of miR-146b suppressed the production of inflammatory factors in the serum of pneumonia patients and WI-38 cells treated with LPS. In addition, elevating miR-146b expression increased WI-38 cell viability and reduced cell apoptosis. More importantly, bioinformatics analysis revealed that MyD88 was a target of miR-146b and could overturn the protective effect of miR-146b on the inflammation injury in LPS-injured WI-38 cells. Furthermore, miR-146b over-expression inhibited the activation of NF-κB signaling pathway by suppressing MyD88.Conclusion: miR-146b attenuated the inflammation injury in pediatric pneumonia through inhibiting MyD88/NF-κB signaling pathway. These preliminarily findings further deepened our understanding of this mechanism and identified new potential therapeutic targets for pediatric pneumonia.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Lili Dong
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Yu Tang
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Min Li
- Department of Respiration, Children's Hospital Affiliated to Zhengzhou University (Zhengzhou Children's Hospital), Zhengzhou City, China
| | - Mingming Zhang
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang City, China
| |
Collapse
|
56
|
Aceves M, Terminel MN, Okoreeh A, Aceves AR, Gong YM, Polanco A, Sohrabji F, Hook MA. Morphine increases macrophages at the lesion site following spinal cord injury: Protective effects of minocycline. Brain Behav Immun 2019; 79:125-138. [PMID: 30684649 DOI: 10.1016/j.bbi.2019.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 01/05/2019] [Accepted: 01/21/2019] [Indexed: 12/13/2022] Open
Abstract
Opioids are among the most effective and widely prescribed medications for the treatment of pain following spinal cord injury (SCI). Spinally-injured patients receive opioids within hours of arrival at the emergency room, and prolonged opioid regimens are often employed for the management of post-SCI chronic pain. However, previous studies in our laboratory suggest that the effects of opioids such as morphine may be altered in the pathophysiological context of neurotrauma. Specifically, we have shown that morphine administration in a rodent model of SCI increases mortality and tissue loss at the injury site, and decreases recovery of motor and sensory function, and overall health, even weeks after treatment. The literature suggests that opioids may produce these adverse effects by acting as endotoxins and increasing glial activation and inflammation. To better understand the effects of morphine following SCI, in this study we used flow cytometry to assess immune-competent cells at the lesion site. We observed a morphine-induced increase in the overall number of CD11b+ cells, with marked effects on microglia, in SCI subjects. Next, to investigate whether this increase in the inflammatory profile is necessary to produce morphine's effects, we challenged morphine treatment with minocycline. We found that pre-treatment with minocycline reduced the morphine-induced increase in microglia at the lesion site. More importantly, minocycline also blocked the adverse effects of morphine on recovery of function without disrupting the analgesic efficacy of this opioid. Together, our findings suggest that following SCI, morphine may exacerbate the inflammatory response, increasing cell death at the lesion site and negatively affecting functional recovery.
Collapse
Affiliation(s)
- Miriam Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Mabel N Terminel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Andre Okoreeh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Alejandro R Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Yan Ming Gong
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Alan Polanco
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| |
Collapse
|
57
|
Woodcock EA, Hillmer AT, Mason GF, Cosgrove KP. Imaging Biomarkers of the Neuroimmune System among Substance Use Disorders: A Systematic Review. MOLECULAR NEUROPSYCHIATRY 2019; 5:125-146. [PMID: 31312635 PMCID: PMC6597912 DOI: 10.1159/000499621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/13/2019] [Indexed: 12/14/2022]
Abstract
There is tremendous interest in the role of the neuroimmune system and inflammatory processes in substance use disorders (SUDs). Imaging biomarkers of the neuroimmune system in vivo provide a vital translational bridge between preclinical and clinical research. Herein, we examine two imaging techniques that measure putative indices of the neuroimmune system and review their application among SUDs. Positron emission tomography (PET) imaging of 18 kDa translocator protein availability is a marker associated with microglia. Proton magnetic resonance spectroscopy quantification of myo-inositol levels is a putative glial marker found in astrocytes. Neuroinflammatory responses are initiated and maintained by microglia and astrocytes, and thus represent important imaging markers. The goal of this review is to summarize neuroimaging findings from the substance use literature that report data using these markers and discuss possible mechanisms of action. The extant literature indicates abused substances exert diverse and complex neuroimmune effects. Moreover, drug effects may change across addiction stages, i.e. the neuroimmune effects of acute drug administration may differ from chronic use. This burgeoning field has considerable potential to improve our understanding and treatment of SUDs. Future research is needed to determine how targeting the neuroimmune system may improve treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Kelly P. Cosgrove
- Departments of Psychiatry, and of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
58
|
Quan B, Zhang H, Xue R. miR-141 alleviates LPS-induced inflammation injury in WI-38 fibroblasts by up-regulation of NOX2. Life Sci 2018; 216:271-278. [PMID: 30500550 DOI: 10.1016/j.lfs.2018.11.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
AIMS The roles of miR-141 in various types of cancers and inflammatory bowel diseases are researched, whereas, little information about its function in lung inflammation is available. This study was designed to explore the effect of miR-141 on inflammation injury in WI-38 cells, possibly providing basis for targeted therapeutic strategy for treatment of infantile pneumonia. MAIN METHODS WI-38 cells were treated with LPS to construct cell model with inflammation injury. Expressions of miR-141 and NOX2 were altered by transfection assay and expressions of them were detected by qRT-PCR or Western blot. Cell viability and apoptosis were evaluated by CCK-8 assay and flow cytometry, respectively. The tested pro-inflammatory factors were analyzed by qRT-PCR and Western blot; and their productions were quantified by ELISA. Main proteins participating in regulation of apoptosis, p38 MAPK pathway and NF-κB pathway were analyzed by Western blot. KEY FINDINGS miR-141 was down-regulated in LPS-treated cells and elevating miR-141 level reduced inflammation extent of WI-38 cells by promoting viability, inhibiting apoptosis, and inhibiting production of tested pro-inflammatory cytokines. NOX2 was up-regulated by miR-141 overexpression. NOX2 silence impaired the cell-protective effect of miR-141. miR-141 inhibited LPS-induced activations of p38 MAPK and NF-κB pathways, which was also mediated by NOX2. SIGNIFICANCE miR-141 alleviated LPS-induced inflammation injury in WI-38 fibroblasts by up-regulating NOX2 and further inhibiting p38 MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Beibei Quan
- Neonatal Ward of Department of Pediatrics, Jining No. 1 People's Hospital, Jining 272011, China
| | - Huating Zhang
- Neonatal Ward of Department of Pediatrics, Jining No. 1 People's Hospital, Jining 272011, China
| | - Ruirui Xue
- Neonatal Ward of Department of Pediatrics, Jining No. 1 People's Hospital, Jining 272011, China.
| |
Collapse
|
59
|
Abstract
This paper is the thirty-ninth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2016 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and CUNY Neuroscience Collaborative, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
60
|
MicroRNA-124 regulates the expression of MEKK3 in the inflammatory pathogenesis of Parkinson's disease. J Neuroinflammation 2018; 15:13. [PMID: 29329581 PMCID: PMC5767033 DOI: 10.1186/s12974-018-1053-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/02/2018] [Indexed: 11/21/2022] Open
Abstract
Background Parkinson’s disease (PD) is the most prevalent neurodegenerative disorder that is characterised by selective loss of midbrain dopaminergic (DA) neurons. Chronic inflammation of the central nervous system is mediated by microglial cells and plays a critical role in the pathological progression of PD. Brain-specific microRNA-124 (miR-124) expression is significantly downregulated in lipopolysaccharide (LPS)-treated BV2 cells and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD. However, whether abnormal miR-124 expression could regulate the activation of microglia remains poorly understood. Methods BV2 cells were activated by exposure to LPS, and the expression levels of miR-124, mitogen-activated protein kinase kinase kinase 3 (MEKK3), and the nuclear factor of kappaB (NF-κB) p-p65 were analysed. Over-expression and knockdown studies of miR-124 were performed to observe the effects on MEKK3/NF-κB signalling pathways, and the induction of pro-inflammatory and neurotoxic factors was assessed. In addition, a luciferase reporter assay was conducted to confirm whether MEKK3 is a direct target of miR-124. Meanwhile, production of miR-124, MEKK3, and p-p65; midbrain DA neuronal death; or activation of microglia were analysed when treated with or without miR-124 in the MPTP-induced model of PD. Results We found that the knockdown of MEKK3 could inhibit the activation of microglia by regulating NF-κB expression. Over-expression of miR-124 could effectively attenuate the LPS-induced expression of pro-inflammatory cytokines and promote the secretion of neuroprotective factors. We also first identified a unique role of miR-124 in mediating the microglial inflammatory response by targeting MEKK3/NF-κB signalling pathways. In the microglial culture supernatant (MCS) transfer model, over-expression of the miR-124 or knockdown of MEKK3 in BV2 cells prevented SH-SY5Y from apoptosis and death. Moreover, MEKK3 and p-p65 were abundantly expressed in the midbrain. Furthermore, their expression levels increased and microglial activation was observed in the MPTP-induced model of PD. In addition, exogenous delivery of miR-124 could suppress MEKK3 and p-p65 expression and attenuate the activation of microglia in the substantia nigra pars compacta of MPTP-treated mice. miR-124 also could prevent MPTP-dependent apoptotic midbrain DA cell death in a MPTP-induced PD model. Conclusions Taken together, our data suggest that miR-124 can inhibit neuroinflammation in the development of PD by regulating the MEKK3/NF-κB signalling pathways and implicate miR-124 as a potential therapeutic target for regulating the inflammatory response in PD.
Collapse
|
61
|
Maduna T, Audouard E, Dembélé D, Mouzaoui N, Reiss D, Massotte D, Gaveriaux-Ruff C. Microglia Express Mu Opioid Receptor: Insights From Transcriptomics and Fluorescent Reporter Mice. Front Psychiatry 2018; 9:726. [PMID: 30662412 PMCID: PMC6328486 DOI: 10.3389/fpsyt.2018.00726] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Microglia activation contributes to chronic pain and to the adverse effects of opiate use such as analgesic tolerance and opioid-induced hyperalgesia. Both mu opioid receptor (MOR) encoded by Oprm1/OPRM1 gene and toll like receptor 4 (TLR4) have been reported to mediate these morphine effects and a current question is whether microglia express the Oprm1 transcript and MOR protein. The aim of this study was to characterize Oprm1-MOR expression in naive murine and human microglia, combining transcriptomics datasets previously published by other groups with our own imaging study using the Cx3cr1-eGFP-MOR-mCherry reporter mouse line. Methods: We analyzed microglial Oprm1/OPRM1 expression obtained from transcriptomics datasets, focusing on ex vivo studies from adult wild-type animals and adult post-mortem human cerebral cortex. Oprm1, as well as co-regulated gene sets were examined. The expression of MOR in microglia was also investigated using our novel fluorescent Cx3cr1-eGFP-MOR-mcherry reporter mouse line. We determined whether CX3cR1-eGFP positive microglial cells expressed MOR-mCherry protein by imaging various brain areas including the Frontal Cortex, Nucleus Accumbens, Ventral Tegmental Area, Central Amygdala, and Periaqueductal Gray matter, as well as spinal cord. Results: Oprm1 expression was found in all 12 microglia datasets from mouse whole brain, in 7 out of 8 from cerebral cortex, 3 out of 4 from hippocampus, 1 out of 1 from striatum, and 4 out of 5 from mouse or rat spinal cord. OPRM1 was expressed in 16 out of 17 microglia transcriptomes from human cerebral cortex. In Cx3cr1-eGFP-MOR-mCherry mice, the percentage of MOR-positive microglial cells ranged between 35.4 and 51.6% in the different brain areas, and between 36.8 and 42.4% in the spinal cord. Conclusion: The comparative analysis of the microglia transcriptomes indicates that Oprm1/OPRM1 transcripts are expressed in microglia. The investigation of Cx3cr1-eGFP-MOR-mCherry mice also shows microglial expression of MOR proteinin the brain and spine. These results corroborate functional studies showing the actions of MOR agonists on microglia and suppression of these effects by MOR-selective antagonists or MOR knockdown.
Collapse
Affiliation(s)
- Tando Maduna
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Emilie Audouard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Doulaye Dembélé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Nejma Mouzaoui
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Dominique Massotte
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
62
|
Doyle HH, Murphy AZ. Sex-dependent influences of morphine and its metabolites on pain sensitivity in the rat. Physiol Behav 2017; 187:32-41. [PMID: 29199028 DOI: 10.1016/j.physbeh.2017.11.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/21/2022]
Abstract
Preclinical studies report that the effective dose for morphine is approximately 2-fold higher in females than males. Following systemic administration, morphine is metabolized via Phase II glucuronidation in the liver and brain into two active metabolites: morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G), each possessing distinct pharmacological profiles. M6G binds to μ opioid receptors and acts as a potent analgesic. In contrast, M3G binds to toll-like receptor 4 (TLR4), initiating a neuroinflammatory response that directly opposes the analgesic effects of morphine and M6G. M3G serum concentrations are 2-fold higher in females than males, however, sex-specific effects of morphine metabolites on analgesia and glial activation in vivo remain unknown. The present studies test the hypothesis that increased M3G, and subsequent TLR4-mediated activation of glia, is a primary mechanism driving the attenuated response to morphine in females. We demonstrate that intra-PAG M6G results in a greater analgesic response in females than morphine alone. M6G analgesia was reversed with co-administration of (-)-naloxone, but not (+)-naloxone, suggesting that this effect is μ opioid receptor mediated. In contrast, intra-PAG administration of M3G significantly attenuated the analgesic effects of systemic morphine in males only, increasing the 50% effective dose of morphine two-fold (5.0 vs 10.3mg/kg) and eliminating the previously observed sex difference. An increase in IL-1β, IL-6 and TNF was observed in females following intra-PAG morphine or M6G. In males, only IL-1β levels increased following morphine. Changes in cytokine levels following M3G were limited to TNF in females. Together, these data implicate sex differences in morphine metabolism, specifically M3G, as a contributing factor in the attenuated response to morphine observed in females.
Collapse
Affiliation(s)
- H H Doyle
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - A Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States.
| |
Collapse
|
63
|
Ma Y, Shi Q, Wang J, Xiao K, Sun J, Lv Y, Guo M, Zhou W, Chen C, Gao C, Zhang BY, Dong XP. Reduction of NF-κB (p65) in Scrapie-Infected Cultured Cells and in the Brains of Scrapie-Infected Rodents. ACS Chem Neurosci 2017; 8:2535-2548. [PMID: 28783945 DOI: 10.1021/acschemneuro.7b00273] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Transcription factor NF-κB functions as a pleiotropic regulator of target genes controlling physiological function as well as pathological processes of many different diseases, including some neurodegenerative diseases. However, the role of NF-κB in the pathogenesis of prion disease remains ambiguous. In this study, the status of NF-κB (p65) in a prion-infected cell line SMB-S15 was first evaluated. Significantly lower levels of p65 and the phosphorylated form of p65 (p-p65) were detected in SMB-S15 cells, compared with its normal partner cell line SMB-PS. Markedly slower responses of the NF-κB system to the stimulation of TNF-α were observed in SMB-S15 cells. Removal of PrPSc replication in SMB-S15 cells rescued the expression and activity of NF-κB. However, overexpression of p65 in SMB-S15 cells did not influence the propagation of PrPSc. Moreover, significant decline of p65 level was also observed in the brain tissues of mice infected with the lysates of SMB-S15 cells and hamsters infected with scrapie agent 263K at terminal stage. Immunofluorescence assays (IFAs) on brain sections from either normal or scrapie-infected rodents revealed colocalization of p65 with neuronal nuclear (NeuN) protein positive cells but not with glial fibrillary acidic protein (GFAP) positive cells. Assays of the agents involving in the regulation of NF-κB showed down-regulated phosphoinositide 3-kinase (PI3K) and protein kinase B (PKB/Akt) both in SMB-S15 cells and in the brains of scrapie-infected rodents. Those data indicate a remarkable repression of the classical NF-κB pathway during prion infection both in vitro and in vivo. The alteration of NF-κB (p65) shows close association with the replication and accumulation of PrPSc in the cells.
Collapse
Affiliation(s)
- Yue Ma
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Jing Wang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Jing Sun
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Yan Lv
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Man Guo
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Wei Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Chen Gao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Bao-Yun Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Chang-Bai Rd 155, Beijing 102206, People’s Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment
of Infectious Diseases, Zhejiang University, Hangzhou, 310003, People’s Republic of China
- Key Laboratory
of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s Republic of China
| |
Collapse
|
64
|
Roeckel LA, Utard V, Reiss D, Mouheiche J, Maurin H, Robé A, Audouard E, Wood JN, Goumon Y, Simonin F, Gaveriaux-Ruff C. Morphine-induced hyperalgesia involves mu opioid receptors and the metabolite morphine-3-glucuronide. Sci Rep 2017; 7:10406. [PMID: 28871199 PMCID: PMC5583172 DOI: 10.1038/s41598-017-11120-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
Opiates are potent analgesics but their clinical use is limited by side effects including analgesic tolerance and opioid-induced hyperalgesia (OIH). The Opiates produce analgesia and other adverse effects through activation of the mu opioid receptor (MOR) encoded by the Oprm1 gene. However, MOR and morphine metabolism involvement in OIH have been little explored. Hence, we examined MOR contribution to OIH by comparing morphine-induced hyperalgesia in wild type (WT) and MOR knockout (KO) mice. We found that repeated morphine administration led to analgesic tolerance and hyperalgesia in WT mice but not in MOR KO mice. The absence of OIH in MOR KO mice was found in both sexes, in two KO global mutant lines, and for mechanical, heat and cold pain modalities. In addition, the morphine metabolite morphine-3beta-D-glucuronide (M3G) elicited hyperalgesia in WT but not in MOR KO animals, as well as in both MOR flox and MOR-Nav1.8 sensory neuron conditional KO mice. M3G displayed significant binding to MOR and G-protein activation when using membranes from MOR-transfected cells or WT mice but not from MOR KO mice. Collectively our results show that MOR is involved in hyperalgesia induced by chronic morphine and its metabolite M3G.
Collapse
Affiliation(s)
- Laurie-Anne Roeckel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Valérie Utard
- Université de Strasbourg, Illkirch, France.,Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Illkirch, France
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Jinane Mouheiche
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Hervé Maurin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Anne Robé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - Emilie Audouard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - John N Wood
- Molecular Nociception group, Wolson Institute for Biomedical Research, University College London, WCIE 6BT, London, UK
| | - Yannick Goumon
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Frédéric Simonin
- Université de Strasbourg, Illkirch, France.,Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Illkirch, France
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France. .,Université de Strasbourg, Illkirch, France. .,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France. .,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.
| |
Collapse
|
65
|
Wang X, Wu Q, Liu A, Anadón A, Rodríguez JL, Martínez-Larrañaga MR, Yuan Z, Martínez MA. Paracetamol: overdose-induced oxidative stress toxicity, metabolism, and protective effects of various compounds in vivo and in vitro. Drug Metab Rev 2017; 49:395-437. [PMID: 28766385 DOI: 10.1080/03602532.2017.1354014] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Paracetamol (APAP) is one of the most widely used and popular over-the-counter analgesic and antipyretic drugs in the world when used at therapeutic doses. APAP overdose can cause severe liver injury, liver necrosis and kidney damage in human beings and animals. Many studies indicate that oxidative stress is involved in the various toxicities associated with APAP, and various antioxidants were evaluated to investigate their protective roles against APAP-induced liver and kidney toxicities. To date, almost no review has addressed the APAP toxicity in relation to oxidative stress. This review updates the research conducted over the past decades into the production of reactive oxygen species (ROS), reactive nitrogen species (RNS), and oxidative stress as a result of APAP treatments, and ultimately their correlation with the toxicity and metabolism of APAP. The metabolism of APAP involves various CYP450 enzymes, through which oxidative stress might occur, and such metabolic factors are reviewed within. The therapeutics of a variety of compounds against APAP-induced organ damage based on their anti-oxidative effects is also discussed, in order to further understand the role of oxidative stress in APAP-induced toxicity. This review will throw new light on the critical roles of oxidative stress in APAP-induced toxicity, as well as on the contradictions and blind spots that still exist in the understanding of APAP toxicity, the cellular effects in terms of organ injury and cell signaling pathways, and finally strategies to help remedy such against oxidative damage.
Collapse
Affiliation(s)
- Xu Wang
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain.,b National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Qinghua Wu
- c College of Life Science , Yangtze University , Jingzhou , China.,d Faculty of Informatics and Management , Center for Basic and Applied Research, University of Hradec Kralove , Hradec Kralove , Czech Republic
| | - Aimei Liu
- b National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Arturo Anadón
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - José-Luis Rodríguez
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - María-Rosa Martínez-Larrañaga
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - Zonghui Yuan
- b National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Huazhong Agricultural University , Wuhan , Hubei , China.,e MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China.,f Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety , Wuhan , Hubei , China
| | - María-Aránzazu Martínez
- a Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| |
Collapse
|
66
|
Liu PW, Yue MX, Zhou R, Niu J, Huang DJ, Xu T, Luo P, Liu XH, Zeng JW. P2Y 12 and P2Y 13 receptors involved in ADPβs induced the release of IL-1β, IL-6 and TNF-α from cultured dorsal horn microglia. J Pain Res 2017; 10:1755-1767. [PMID: 28794655 PMCID: PMC5536317 DOI: 10.2147/jpr.s137131] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective P2 receptors have been implicated in the release of neurotransmitter and pro-inflammatory cytokines due to their response to neuroexcitatory substances in the microglia. Dorsal horn P2Y12 and P2Y13 receptors are involved in the development of pain behavior induced by peripheral nerve injury. However, it is not known whether P2Y12 and P2Y13 receptors activation is associated with the expression and the release of interleukin-1B (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) in cultured dorsal spinal cord microglia. For this reason, we examined the effects of ADPβs (ADP analog) on the expression and the release of IL-1β, IL-6, and TNF-α. Methods and results In this study, we observed the effect of P2Y receptor agonist ADPβs on the expression and release of IL-1β, IL-6 and TNF-α by using real-time fluorescence quantitative polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). ADPβs induced the increased expression of Iba-1, IL-1β, IL-6 and TNF-α at the level of messenger RNA (mRNA). ADPβs-evoked increase in Iba-1, IL-1β, IL-6 and TNF-α mRNA expression was inhibited only partially by P2Y12 receptor antagonist MRS2395 or P2Y13 receptor antagonist MRS2211, respectively. Similarly, ADPβs-evoked release of IL-1β, IL-6 and TNF-α was inhibited only partially by MRS2395 or MRS2211. Furthermore, ADPβs-evoked increased expression of Iba-1, IL-1β, IL-6 and TNF-α mRNA, and release of IL-1β, IL-6 and TNF-α were nearly all blocked after co-administration of MRS2395 plus MRS2179. Further evidence indicated that P2Y12 and P2Y13 receptor-evoked increased gene expression of IL-1β, IL-6 and TNF-α were inhibited by Y-27632 (ROCK inhibitor), SB203580 (P38MAPK inhibitor) and PDTC (NF-κb inhibitor), respectively. Subsequently, P2Y12 and P2Y13 receptor-evoked release of IL-1β, IL-6 and TNF-α, were also inhibited by Y-27632, SB203580 and PDTC, respectively. Conclusion These observations suggest that P2Y12 and P2Y13 receptor-evoked gene expression and release of IL-1β, IL-6 and TNF-α are associated with ROCK/P38MAPK/NF-κb signaling pathway.
Collapse
Affiliation(s)
- Pei-Wen Liu
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Ming-Xia Yue
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Rui Zhou
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Juan Niu
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Du-Juan Huang
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Tao Xu
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Pei Luo
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Xiao-Hong Liu
- Department of Physiology, Zunyi Medical College, Guizhou, China
| | - Jun-Wei Zeng
- Department of Physiology, Zunyi Medical College, Guizhou, China
| |
Collapse
|
67
|
Montesinos J, Gil A, Guerri C. Nalmefene Prevents Alcohol-Induced Neuroinflammation and Alcohol Drinking Preference in Adolescent Female Mice: Role of TLR4. Alcohol Clin Exp Res 2017; 41:1257-1270. [DOI: 10.1111/acer.13416] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/04/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol; Príncipe Felipe Research Center; Valencia Spain
| | - Anabel Gil
- Department of Molecular and Cellular Pathology of Alcohol; Príncipe Felipe Research Center; Valencia Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol; Príncipe Felipe Research Center; Valencia Spain
| |
Collapse
|
68
|
Popiolek-Barczyk K, Piotrowska A, Makuch W, Mika J. Biphalin, a Dimeric Enkephalin, Alleviates LPS-Induced Activation in Rat Primary Microglial Cultures in Opioid Receptor-Dependent and Receptor-Independent Manners. Neural Plast 2017; 2017:3829472. [PMID: 28573049 PMCID: PMC5442438 DOI: 10.1155/2017/3829472] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/12/2017] [Accepted: 04/03/2017] [Indexed: 01/23/2023] Open
Abstract
Neuropathic pain is relatively less responsive to opioids than other types of pain, which is possibly due to a disrupted opioid system partially caused by the profound microglial cell activation that underlines neuroinflammation. We demonstrated that intrathecally injected biphalin, a dimeric enkephalin analog, diminished symptoms of neuropathy in a preclinical model of neuropathic pain in rats (CCI, chronic constriction injury of the sciatic nerve) at day 12 postinjury. Using primary microglial cell cultures, we revealed that biphalin did not influence cell viability but diminished NO production and expression of Iba1 in LPS-stimulated cells. Biphalin also diminished MOP receptor level, as well as pronociceptive mediators (iNOS, IL-1β, and IL-18) in an opioid receptor-dependent manner, and it was correlated with diminished p-NF-κB, p-IκB, p-p38MAPK, and TRIF levels. Biphalin reduced IL-6, IL-10, TNFα, p-STAT3, and p-ERK1/2 and upregulated SOCS3, TLR4, and MyD88; however, this effect was not reversed by naloxone pretreatment. Our study provides evidence that biphalin diminishes neuropathy symptoms, which might be partially related to reduced pronociceptive mediators released by activated microglia. Biphalin may be a putative drug for future pain therapy, especially for the treatment of neuropathic pain, when the lower analgesic effects of morphine are correlated with profound microglial cell activation.
Collapse
Affiliation(s)
| | - Anna Piotrowska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
69
|
Yang WS, Kim D, Yi YS, Kim JH, Jeong HY, Hwang K, Kim JH, Park J, Cho JY. AKT-targeted anti-inflammatory activity of the methanol extract of Chrysanthemum indicum var. albescens. JOURNAL OF ETHNOPHARMACOLOGY 2017; 201:82-90. [PMID: 28274893 DOI: 10.1016/j.jep.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wild chrysanthemum (Chrysanthemum indicum) is one of well-known medicinal plants traditionally used in Korea and China. As a variant of wild chrysanthemum, white wild chrysanthemum (Chrysanthemum indicum var. albescens) is also ethnopharmacologically applied to treat various symptoms such as inflammatory diseases. AIM OF STUDY Although the anti-inflammatory activity of Chrysanthemum indicum has been reported, the anti-inflammatory activity and underlying molecular mechanism of white wild chrysanthemum are poorly understood. MATERIALS AND METHODS The effects of Chrysanthemum indicum var. albescens methanol extract (Civ-ME) on the production of inflammatory mediators, expression of pro-inflammatory genes, cell viability, and the activities of intracellular signaling molecules and transcription factors were investigated in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. RESULTS Civ-ME suppressed the production of both nitric oxide (NO) and prostaglandin E2 (PGE2) without cytotoxicity in LPS-stimulated RAW264.7 cells. Civ-ME was found to reduce the mRNA levels of inflammatory genes such as inducible NO synthase (iNOS) and tumor necrosis factor (TNF)-α and reduced NF-κB-mediated transcriptional activation. Civ-ME inhibited the nuclear translocation of NF-κB (p65 and p50), and its upstream signaling composed of IκBα and IKKα/β. An NF-κB luciferase reporter gene assay and an in vitro kinase assay confirmed that AKT1 and AKT2 might be direct pharmacological targets of Civ-ME. In addition, luteolin was identified by HPLC analysis as the main active pharmacological components of Civ-ME. CONCLUSION Civ-ME exerts an anti-inflammatory effect by targeting AKT1 and AKT2 in the NF-κB signaling pathway in macrophage-mediated inflammatory responses.
Collapse
Affiliation(s)
- Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Donghyun Kim
- Heritage Material Research Team, Amorepacific R&D Unit, Yongin 446-729, Republic of Korea.
| | - Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28503, Republic of Korea.
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Hye Yoon Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Kyeonghwan Hwang
- Heritage Material Research Team, Amorepacific R&D Unit, Yongin 446-729, Republic of Korea.
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| | - Junseong Park
- Heritage Material Research Team, Amorepacific R&D Unit, Yongin 446-729, Republic of Korea.
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
70
|
A 2B adenosine receptors stimulate IL-6 production in primary murine microglia through p38 MAPK kinase pathway. Pharmacol Res 2016; 117:9-19. [PMID: 27974241 DOI: 10.1016/j.phrs.2016.11.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/17/2016] [Accepted: 11/20/2016] [Indexed: 01/22/2023]
Abstract
The hallmark of neuroinflammation is the activation of microglia, the immunocompetent cells of the CNS, releasing a number of proinflammatory mediators implicated in the pathogenesis of neuronal diseases. Adenosine is an ubiquitous autacoid regulating several microglia functions through four receptor subtypes named A1, A2A, A2B and A3 (ARs), that represent good targets to suppress inflammation occurring in CNS. Here we investigated the potential role of ARs in the modulation of IL-6 secretion and cell proliferation in primary microglial cells. The A2BAR agonist 2-[[6-Amino-3,5-dicyano-4-[4-(cyclopropylmethoxy)phenyl]-2-pyridinyl]thio]-acetamide (BAY60-6583) stimulated IL-6 increase under normoxia and hypoxia, in a dose- and time-dependent way. In cells incubated with the blockers of phospholipase C (PLC), protein kinase C epsilon (PKC-ε) and PKC delta (PKC-δ) the IL-6 increase due to A2BAR activation was strongly reduced, whilst it was not affected by the inhibitor of adenylyl cyclase (AC). Investigation of cellular signalling involved in the A2BAR effect revealed that only the inhibitor of p38 mitogen activated protein kinase (MAPK) was able to block the agonist's effect on IL-6 secretion, whilst inhibitors of pERK1/2, JNK1/2 MAPKs and Akt were not. Stimulation of p38 by BAY60-6583 was A2BAR-dependent, through a pathway affecting PLC, PKC-ε and PKC-δ but not AC, in both normoxia and hypoxia. Finally, BAY60-6583 increased microglial cell proliferation involving A2BAR, PLC, PKC-ε, PKC-δ and p38 signalling. In conclusion, A2BARs activation increased IL-6 secretion and cell proliferation in murine primary microglial cells, through PLC, PKC-ε, PKC-δ and p38 pathways, thus suggesting their involvement in microglial activation and neuroinflammation.
Collapse
|