Page 5 - Read Online
P. 5

Davis et al. Neuroimmunol Neuroinflammation 2020;7:300-10          Neuroimmunology
               DOI: 10.20517/2347-8659.2020.19                              and Neuroinflammation




               Original Article                                                              Open Access


               β-funaltrexamine differentially modulates chemokine
               and cytokine expression in normal human

               astrocytes and C20 human microglial cells


               Randall L. Davis, Kelly McCracken, Daniel J. Buck

               Department of Pharmacology/Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK 74107, USA.

               Correspondence to: Dr. Randall L. Davis, Department of Pharmacology/Physiology, Oklahoma State University Center for Health
               Sciences, 1111 West 17th Street, Tulsa, OK 74107, USA. E-mail: randall.davis@okstate.edu

               How to cite this article: Davis RL, McCracken K, Buck DJ. β-funaltrexamine differentially modulates chemokine and cytokine
               expression in normal human astrocytes and C20 human microglial cells. Neuroimmunol Neuroinflammation 2020;7:300-10
               http://dx.doi.org/10.20517/2347-8659.2020.19

               Received: 26 Feb 2020    First Decision: 27 May 2020    Revised: 4 Jun 2020    Accepted: 17 Jun 2020    Available online: 15 Aug 2020
               Science Editor: Athanassios P. Kyritsis    Copy Editor: Cai-Hong Wang    Production Editor: Jing Yu



               Abstract
               Aim: Emerging evidence implicates astrocyte/microglia dysregulation in a range of brain disorders, thereby making
               glial cells potential therapeutic targets. The novel anti-inflammatory actions of beta-funaltrexamine (β-FNA) are of
               particular interest. β-FNA is a derivative of naltrexone, and recognized as a selective, irreversible antagonist at the
               mu-opioid receptor (MOR). However, we discovered that β-FNA has novel anti-inflammatory actions that seem to
               be mediated through a MOR-independent mechanism. Thus far, we have focused on the acute effects of β-FNA on
               inflammatory signaling.


               Methods: The effect of β-FNA treatment on interleukin-1β (IL-1β)-induced inflammatory signaling in normal human
               astrocytes (NHA) and C20 human microglial cells. Cytokine/chemokine expression was measured using ELISA,
               and nuclear factor-kappaB (NF-κB) p65 activation was evaluated by immunoblot.


               Results: IL-1β-induced interferon-γ inducible protein-10 (CXCL10) production in NHA was more sensitive to chronic
               (3 day) β-FNA as indicated by an approximately 3-fold lower EC 50  compared to that observed in acutely treated
               cells. Chronic β-FNA did not affect IL-1β-induced monocyte chemoattractant protein-1 (CCL2) or IL-6 production
               in NHA. β-FNA inhibited phosphorylation of NF-κB p65, suggesting that the inhibitory effects may be due in part
               to reduced NF-κB activation. We showed for the first time that C20 human microglial cells were insensitive to the
               anti-inflammatory actions of acute β-FNA.



                           © The Author(s) 2020. Open Access This article is licensed under a Creative Commons Attribution 4.0
                           International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use,
                sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long
                as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license,
                and indicate if changes were made.


                                                                                                                                                       www.nnjournal.net
   1   2   3   4   5   6   7   8   9   10