Page 265 - Read Online
P. 265

Burns et al.                                                                                                                                                                        Altered filamin A enables Aβ signaling

                                                              has not been elucidated. The tau proteopathy in AD,
                                                              therefore, involves hyperphosphorylation, but may or
                                                              may not include misfolding. The formation of PHFs
                                                              requires hyperphosphorylated tau, and tau protein in
                                                              neurofibrillary tangles is hyperphosphorylated, most
                                                              notably at Ser 202 , Thr 231  and Thr 181[41] . Interestingly,
                                                              the alpha-synuclein that forms fibrils and is abundant
                                                              in Lewy bodies in Parkinson’s disease is also
                                                              hyperphosphorylated, at a single serine site [42] .


                                                              ALTERED FLNA LINKS Aβ AND TAU
                                                              PROTEOPATHIES

                                                              We recently described a third, atypical proteopathy in
                                                              AD that is critically interconnected with the toxicities
                                                              of both Aβ 42  and tau. This third proteopathy is an
                                                              altered conformation of the scaffolding protein FLNA.
                                                              It is induced by Aβ 42 , and in reciprocal action, enables
                                                              Aβ 42 ’s toxic signaling via α7nAChR to activate kinases
                                                              that hyperphosphorylate tau [13] . Altered FLNA enables
              N                  N                            Aβ 42 ’s signaling via α7nAChR by associating with this
           Figure 1: Altered FLNA linkage to α7nAChR enables Aβ 42 ’s toxic   receptor [13,15] . Although FLNA constitutively associates
           signaling via α7nAChR to hyperphosphorylate tau. Monomers or   with other receptors including the mu opioid receptor
           small oligomers of Aβ 42  bind α7nAChR, which recruits FLNA to link   and insulin receptors [43] , FLNA does not normally link
           to α7nAChR. This recruitment likely alters FLNA’s conformation,
           which in turn increases the affinity of the Aβ 42 -α7nAChR   to α7nAChR. We hypothesize that upon Aβ 42  binding
           interaction to a femtomolar affinity and enables the signaling.   to α7nAChR, FLNA is recruited to this receptor and
           ERK1 and JNK kinases are activated to hyperphosphorylate   its conformation altered to enable Aβ 42 ’s aberrant
           tau. Hyperphosphorylated tau loses its function of stabilizing
           microtubules and dissociates from them, eventually creating PHFs   signaling [Figure 1]. FLNA in control brains can be
           and neurofibrillary tangles. FLNA: filamin A; Aβ: amyloid beta;   induced to link to α7nAChR by incubation with Aβ 42  in
           α7nAChR: α7 nicotinic acetylcholine receptor; PHF: paired helical   vitro or by ICV Aβ 42  infusion in vivo [13,15] . The altered
           filament
                                                              conformation of FLNA is also illustrated by the 100-
                                                              fold difference in binding affinities of the novel drug
           soluble Aβ 42 , their nanomolar or lower affinities   candidate PTI-125 to FLNA immunopurified from
           suggest high off-rates and limited target engagement,   human postmortem AD vs. control brain [13] . One
           in contrast to Aβ 42 ’s nearly irreversible sub-picomolar   distinct difference between altered FLNA and other
           interaction with α7nAChR. Other targets of soluble Aβ   proteopathies is that the altered conformation of FLNA
           include PrP , a prion receptor, which Aβ binds at 50-  does not appear to promote self-aggregation.
                      C
           100 nmol/L to suppress LTP in slice cultures [37] . Acting
                                         c
           as a co-receptor for the Aβ-PrP  complex, mGluR5   Best known for cross-linking actin to enable cell
           also plays a role in the impaired LTP [38] . Soluble Aβ   structure, flexibility and motility, FLNA is a prominent
           has also been shown to bind neuroligin-1, a post-  regulator of the actin cytoskeletal assembly and
           synaptic cell adhesion protein, in the nanomolar range,   dynamics [44-46] .  The  actin  cytoskeleton,  a  vital
           and this interaction has been proposed to promote Aβ   component in synapses and the dendritic network,
           oligomer formation [39] .                          is impaired in AD [47] . Hence, the FLNA proteopathy
                                                              might also disrupt synaptic and dendritic function
           Evidence  that  phosphorylation can  alter tau’s   in AD by disrupting actin dynamics. FLNA exists
           conformation comes from a study showing that a     as an intracellular homodimer and dimerizesin a
           particular AD-specific phosphorylated tau species   membrane-bound, C-terminal domain. It is a large
           is only formed when specific phosphoepitopes in a   (280-kDa) protein with 24 immunoglobulin repeats
           proline-rich region are sequentially phosphorylated by   that are natively β-sheet pleated, forming two rod-like
           GSK-3β (at Thr212) and then by PKA (at Ser214) [40] .   domains separated by two hinge regions. The nature
           Whether  tau’s  hyperphosporylation  contributes   of the conformational change of FLNA in AD is not
           to protein misfolding prior to formation of PHFs is   yet known, thoughit is interesting that native FLNA is
           speculative, particularly as its pathological structure   predominantly β-sheeted. Demonstrating induction by

                          Neuroimmunology and Neuroinflammation ¦ Volume 4 ¦ December 8, 2017             265
   260   261   262   263   264   265   266   267   268   269   270