Page 264 - Read Online
P. 264

Burns et al.                                                                                                                                                                        Altered filamin A enables Aβ signaling

           ultimately form the histopathological hallmarks of AD   described proteopathy is an altered conformation of
           brain: amyloid plaques and tau-containing tangles.   the ubiquitous scaffolding protein filamin A (FLNA),
           Misfolded dysfunctional proteins and their aggregation   induced by Aβ 42  and without apparent aggregation [13] .
           occurs in many other neurodegenerative diseases    The 1st toxic cascade of Aβ enabled by altered
           including Parkinson’s disease, dementia with Lewy   FLNA is Aβ’s signaling via α7 nicotinic acetylcholine
           bodies, multiple systems atrophy, frontotemporal   receptor (α7nAChR) to hyperphosphorylate tau. The
           dementia, amyotrophic lateral sclerosis and        2nd signaling pathway is Aβ’s aberrant activation of
           Huntington’s disease [1-4] . Typically, misfolded proteins   toll-like-receptor 4 (TLR4), by binding CD14 [14] , to
           self-aggregate, creating intracellular inclusions that   induce neuroinflammation. In both cases, altered
           become extracellular deposits following cell death. In   FLNA associates with these receptors to allow their
           many cases, misfolded proteins propagate in a cell-to-  aberrant signaling by Aβ [13,15] . Native FLNA in control
           cell “prion-like” manner [1-3,5] .                 brains does not associate with either receptor.
                                                              Whether the altered conformation precedes or is a
           The proteopathy of Aβ in AD is an amyloidosis,     consequence of these receptor linkages is discussed.
           meaning the protein converts from an α-helix-rich   The  interdependence  of  altered  FLNA  with  Aβ
           state to a β-sheet conformation. To enter an amyloid   signaling to hyperphosphorylate tau and promote
           or β-sheet state, proteins must expose the backbone   neuroinflammation has been elucidated via the
           amide  N-H  and  C=O  groups  to  allow  hydrogen   reversal of the FLNA proteopathy by a small molecule
                  [4]
           bonding . Cleavage of amyloid precursor protein into   therapeutic candidate, PTI-125.
           Aβ 42  by secretases can expose these amide N-H and
                                                         [4]
           C=O groups and promote a β-sheet conformation .    Aβ SIGNALING VIA α7NACHR TO
           Elevated concentrations from overproduction or     HYPERPHOSPHORYLATE TAU
           insufficient clearance/processing may also contribute.
           The  hydrogen  bonding  of  the  pleated,  β-sheet   The most toxic form of Aβ is considered soluble Aβ
           conformation between parallel or anti-parallel β-sheets   oligomers rather than plaque deposits [16,17] . Evidence
           is much stronger than that in native α-helices, making   that soluble Aβ induces tau pathology has grown, with
           reversal unlikely. Additionally, the β-sheet conformation   hyperphosphorylation as the primary pathological
           allows hydrogen bonding between separate, stacked   modification [18] . Extensive research has elucidated
           molecules, promoting oligomerization and eventual   the role of the α7nAChR in the toxicity of soluble Aβ 42
                           [4]
           plaque formation . Aβ is proposed to form a toxic,   and the consequent hyperphosphorylation of tau [19-24] .
           small oligomer “seed” requiring 3 or 4 molecules   Soluble Aβ 42  in monomeric or oligomeric form binds
           used as a template to “infect” native molecules and   and signals via α7nAChR [25-28] , essentially hijacking this
           propagate in a prion-like manner .                 receptor to abnormally activate various kinases [27,29-31]
                                        [6]
                                                              to heighten tau phosphorylation. Supportive data
           The proteopathy of tau, or tauopathy, in AD is primarily   include co-localization of Aβ 42  and α7nAChR in AD
           caused by hyperphosphorylation. Hyperphosphorylated   pyramidal neurons and a complete blockade of Aβ 42 -
           tau loses its function of stabilizing microtubules and   induced tau hyperphosphorylation in vitro by α7nAChR
                                [7]
           dissociates from them . The increased pool of free   antisense oligonucleotides [32] . Aβ 42  dose-dependently
           tau after dissociation from microtubules is likely an   activates tau kinases to persistently phosphorylate tau
                                               [8]
           important first step to aggregation in AD . Untethered   at the three proline-directed sites, resulting in elevated
           from microtubules, hyperphosphorylated tau twists   hyperphosphorylated tau in neurofibrillary tangles.
           together to form the paired helical filaments (PHFs)   This Aβ-driven tau hyperphosphorylation can also be
           found in neurofibrillary tangles. In a toxic gain of   blocked by the α7nAChR antagonist α-bungarotoxin
           function, hyperphosphorylated tau also actively disrupts   or  other  α7nAChR  ligands  if  administered
           microtubules and inhibits their assembly [7,9]  and even   prophylactically [32-35] . The hyperphosphorylation of tau
           sequesters functional tau and other microtubule    renders it dysfunctional, alters its cellular distribution
                              [9]
           associated proteins . Hyperphosphorylation also    and disrupts axonal/dendritic transport, leading to
           changes tau’s localization from axon-predominant to   neurofibrillary lesions, dendritic breakdown, and
           include dendrites, neuronal cell bodies and presynaptic   ultimately, neurofibrillary tangles [27] . Importantly,
           areas, leading to synaptic dsyfunction [10-12] .   soluble Aβ 42  binds α7nAChR with an extraordinarily
                                                              high (high femtomolar) affinity, rendering the Aβ 42 -
           This mini-review focuses on a third, interconnected   α7nAChR interaction nearly irreversible [26,36] .
           proteopathy in AD and its critical role in 2 toxic
           signaling  pathways  of  soluble  Aβ.  The  newly   Though other targets have been demonstrated for

            264                                                              Neuroimmunology and Neuroinflammation ¦ Volume 4 ¦ December 8, 2017
   259   260   261   262   263   264   265   266   267   268   269