Page 6 - Read Online
P. 6

Davis et al. Neuroimmunol Neuroinflammation 2020;7:300-10  I  http://dx.doi.org/10.20517/2347-8659.2020.19            Page 301

               Conclusion: β-FNA differentially affects inflammatory cytokine/chemokine expression in human astrocytes and
               microglia. These findings warrant further investigation into the novel anti-inflammatory actions of β-FNA, with
               a particular focus on astrocytes. These insights should contribute to the development of strategies to treat brain
               disorders that involve neuroinflammation.

               Keywords: Neuroinflammation, brain, opioid, nuclear factor-kappaB, pro-inflammatory





               INTRODUCTION
               Neuroinflammation is present in brain infection, trauma, neurodegenerative diseases, and psychiatric
                             [1-5]
               disorders as well . Glial cells, including astrocytes and microglia are instrumental in neuroinflammation.
               The therapeutic effectiveness of certain antidepressant and antipsychotic drugs reportedly results, in part,
                                          [6-9]
               from anti-inflammatory actions . Astrocytes and microglia have integral roles in metabolic/neurotrophic
               support, ion and neurotransmitter homeostasis, synaptic plasticity, and modulation of neuronal excitability;
               and they respond to infection and cellular insults associated with neuropathology [10-13] . Astrocytes have
               a fundamental role in neuropathogenesis, in part, through the release of neurotoxic/neuroinflammatory
               factors including cytokines [i.e., interleukin (IL)-1β and IL-6] and chemokines [interferon-γ inducible
               protein-10 (CXCL10) and monocyte chemoattractant protein-1 (CCL2)]. These secreted factors can be
               directly neurotoxic or can induce further glial activation/dysregulation [14-17] . Microglia are resident CNS
               macrophages and thus play an important role in neuroinflammation, in part, through phagocytosis of
               cellular debris and release of inflammatory mediators including cytokines/chemokines. Importantly,
               emerging evidence highlights the importance of astrocyte/microglia dysregulation in a wide range of CNS
               disorders, thus implicating glial cells as potential therapeutic targets [18-21] .


               We, and others, are therefore interested in identifying novel, anti-inflammatory agents that are
               therapeutically effective in the treatment of neurological disorders. We are interested in the previously
               identified, novel anti-inflammatory actions of beta-funaltrexamine (β-FNA). As a fumaramate methyl
               ester derivative of naltrexone, β-FNA is most notably recognized as a selective, irreversible antagonist
               at the mu-opioid receptor (MOR) [22,23] . In both behavioral and in vitro assays, β-FNA acts initially as a
               reversible kappa-opioid receptor agonist, and then later results in MOR antagonism [24,25] . As an alkylating
                                                                                               [23]
               agent, β-FNA irreversibly antagonizes MOR by covalently binding at Lys233 on the receptor . However,
               we discovered that β-FNA also has novel anti-inflammatory actions which seem to be mediated through
               MOR-independent actions [26-28] . For instance, neither naltrexone (a nonselective opioid receptor antagonist)
               nor D-Phe-Cys-Tyr-D-Trp-D-Arg-Pen-Thr-NH2 (CTAP) inhibits pro-inflammatory-induced CXCL10
               expression in human astroglial cells [27,28] . Additionally, we predicted that if the anti-inflammatory actions of
               β-FNA were due to alkylation, this covalent modification should then remain after washout. Indeed, pre-
               treatment of astroglial cells with β-FNA for 60 min, followed by drug washout prior to stimulating with
               IL-1β (or tumor necrosis factor α), resulted in inhibition similar to 24 h co-exposure (cytokine stimulus
               + β-FNA). These findings suggested that β-FNA-induced modifications (i.e., alkylation) are persistent
               and lead to the disruption of signal transduction. Notably, our in vitro findings also showed that β-FNA
               reduces inflammatory signaling in astroglia, regardless of whether the stimulus is tumor necrosis factor α,
               IL-1β or bacterial lipopolysaccharide (LPS). We also determined that β-FNA inhibits LPS-induced pro-
               inflammatory cytokine expression in mouse brain (but not in plasma) . Furthermore, treatment with
                                                                             [29]
                                                                                                      [29]
               β-FNA reduced LPS-induced sickness behavior in mice suggesting important translational implications .
               The primary goal of the present study was to determine the effect of chronic β-FNA treatment on
               inflammatory signaling in NHA. Additionally, we assessed for the first time the effects of  β-FNA on
               inflammatory signaling in human microglial cells.
   1   2   3   4   5   6   7   8   9   10   11