libro abstract - amazon web services · !8! &...

27
!"#$%&#" ("#")%(*)+!" ,+- !"#$%&'(&) +',&-,&. /%'0% -)& 1%& 0%-##&23&, /& 2&&4 15 6-0&78 !"#$ &'()* $+*,'-)($( .# /+-+0* '-1 ."'-(2'0#-'2 '(3+*.( 9'#-25: 9-; <= 1% >=<? 4-)5+"().6 1+/2) 7.81) 1) 9)2'-# :82' ;'3#2+#-)*'< =)' 7'-.>:-.#-)# ?@ @)3-2'$&),. 9A B) C8*'< !A D'"1#-)< :A E#3'-)< 9A 7#"0-#

Upload: lytuyen

Post on 16-Feb-2019

216 views

Category:

Documents


0 download

TRANSCRIPT

 

!"#$%&#"'("#")%(*)+!"',+-''

!"#$%&'(&)*+',&-,&.*/%'0%*-)&*1%&*0%-##&23&,*/&*2&&4*15*6-0&78*!"#$%&'()*%$+*,'-)($(%.#%/+-+0*%'-1%."'-(2'0#-'2%'(3+*.(%

%

9'#-25:*9-;*<=1%*>=<?*

4-)5+"().6%1+/2)%7.81)%1)%9)2'-#%%:82'%;'3#2+#-)*'<%=)'%7'-.>:-.#-)#%?@%

@)3-2'$&),.%9A%B)%C8*'<%!A%D'"1#-)<%:A%E#3'-)<%9A%7#"0-#%

  2  

FINAL  PROGRAMME      

10.00       Opening          

Topic  1  -­‐  Basic  mechanisms    10.15       Keynote  lecturer:    Claudia  Bagni         “Study  of  APP  local  translation  in  physiological  and  pathological  conditions”            11.00           Sarah  Beggiato    

“CHF5074  restores  visual  memory  ability  and  rescue  synaptic  dysfunction  in  pre-­‐plaque  Tg2576  mice”      

11.20     Isabella  Colombo    “JNK  mediated  Aβ  oligomers  synaptic  dysfunction”        

11.40       Coffee  break          

Topic  2  -­‐  Genetic  aspects    

12.00       Keynote  lecturer:  Sandro  Sorbi         “Genetic  of  Alzheimer’s  disease”          12.45         Dmitry  Lim    

“Amyloid   beta   deregulates   astroglial   mGluR5-­‐mediated   Ca2+   signaling   via  calcineurin  and  NF-­‐kB”      

13.10     Massimo  Grilli    “β-­‐amyloid   differently   regulates   the   function   of   nAChRs   which   stimulate   the  release  of  glutamate,  aspartate,  glycine  and  GABA  in  rat  hippocampus”      

13.30       Lunch  and  Poster  Session        

  3  

 Topic  3  -­‐  Translational  aspects  

 15.00       Keynote  lecturer:    Alessandro  Padovani         “Biomarkers,  where  are  we?”      15.45       Valeria  Bortolotto    

“HMGB-­‐1  protein  and  β-­‐amyloid  oligomers  promote  neuronal  differentiation  of  adult   hippocampal   neural   progenitors   via   RAGE/NF-­‐κB   axis:   relevance   for  Alzheimer’s  Disease”      

 16.10     Fabrizio  Piazza    

“Pathogenetic   role   of   CSF   anti-­‐Aβ   autoantibodies   in   CAA-­‐related   inflammation  and   Alzheimer’s   disease:   a   novel   biomarker   for   the   Amyloid   Related   Imaging  Abnormalities  for  amyloid-­‐modifying  therapies”    

   

16.30       Conclusions      

  4  

Index  of  abstracts    

• Biliverdin   Reductase-­‐A:   a   Novel   Drug   Target   for   Atorvastatin   in   a   Dog   Preclinical  Model   of   Alzheimer   Disease   -­‐  Barone  Eugenio,  Head  Elizabeth  and  Butterfield  D.  Allan………………………………………………………...................................6  

 • CHF5074   restores   visual   memory   ability   and   rescue   synaptic   dysfunction   in   pre-­‐plaque   Tg2576  mice   -­‐   Beggiato  

Sarah,  Giuliani  Alessandro,  Baldassarro  Vito  Antonio,  Mangano  Chiara,    Giardino   Luciana,   Antonelli   Tiziana,   Calzà  Laura,  Ferraro  Luca,  Imbimbo  Bruno  Pietro……………………………………………………………………………………………………………..7  

 • HMGB-­‐1   protein   and   β-­‐amyloid   oligomers   promote   neuronal   differentiation   of   adult   hippocampal   neural  

progenitors   via   RAGE/NF-­‐κB   axis:   relevance   for   Alzheimer’s   Disease   -­‐   Bortolotto   Valeria,   Meneghini   Vasco,  Francese   Maria   Teresa,   Dellarole   Anna,   Carraro   Lorenzo,   Terzieva   Slavica   and   Grilli  Mariagrazia……………………………………………………………………………………………………………………………………………………………..8    

• JNK   mediated   Aβ   oligomers   synaptic   dysfunction   -­‐   Colombo   I.,   Sclip   A.,   Veglianese   P.,   Messa   M.,   Colombo   L.,  Salmona  M.  and  Borsello  T………………………………………………………………………………………….…………………………………………..9  

 • Role  of  donepezil  in  influencing  endogenous  immune  response  against  Abeta,  an  ex  vivo  and  in  vitro  study  -­‐  Conti  

Elisa,   Tremolizzo   Lucio,   Santarone   Marta   Elena,   Tironi   Marco,   Zoia   Chiara   Paola,   Appollonio   Ildebrando   and  Ferrarese  Carlo………………………………………………………………………………………………….…………………………………………………..10  

 • Acety-­‐L-­‐carnitine  is  a  potent  positive  modulator  of  adult  hippocampal  neurogenesis  in  vitro  and  in  vivo  -­‐  Cuccurazzu  

Bruna,   Bortolotto   Valeria,   Valente   Maria   Maddalena,   Ubezio   Federica,   Canonico   Pier   Luigi   and   Grilli  Mariagrazia…………………………………………………………………………………………………………………………..……………………………….11  

 • Neuroprotection   by   association   of   palmitoylethanolamide   with   luteolin   (Ġlìalia®)   in   experimental   Alzheimer’s  

disease   models:     the   control   of   neuroinflammation   -­‐   Esposito   Emanuela,   Paterniti   Irene,   Campolo  Michela   and  Cuzzocrea  Salvatore………………………………………………………………………………………………………………………………….……………12  

 • SUMOylation/deSUMOylation:  a  balance  to  be  re-­‐equilibrated  -­‐  Feligioni  Marco  and  Nisticò  Robert…………………….13  

 • β-­‐amyloid  differently  regulates  the  function  of  nAChRs  which  stimulate  the  release  of  glutamate,  aspartate,  glycine  

and  GABA  in  rat  hippocampus  -­‐  Grilli  Massimo,  Zappettini  Stefania,  Mura  Elisa,  Preda  Stefania,  Salamone  Alessia,  Olivero  Guiendalina,  Govoni  Stefano  and  Marchi  Mario…………………………………………………………………………………………14  

 • Amyloid-­‐b  and  Alzheimer´s  disease  type  pathology  differentially  affects  calcium  signalling  toolkit  in  astrocytes  from  

different  brain  regions  -­‐  Grolla  Ambra  A,  Sim  Joan  A,  Lim  Dmitry,  Rodriguez  Jose  Julio,  Genazzani  Armando  A  and  Verkhratsky  Alexei…………………………………………………………………………………………………………………………………………………15  

 • Disease   modifying   effects   of   the   polyphenol   Oleuropein   Aglycone   on   Alzheimer’s   Disease:   a   Preclinical   Study   -­‐  

Grossi  Cristina,  Rigacci  Stefania,  Ed  Dami  Teresa,  Luccarini  Ilaria,  Stefani  Massimo  and  Casamenti  Fiorella…………..16    

• Bv8/prokineticin  2   is   a  potential  mediator  of  Alzheimer’s  disease   -­‐  Lattanzi  Roberta,   Severini  Cinzia,  Ciotti  Maria  Teresa,  Petrocchi  Pamela,  Marconi  Veronica,  Giancotti  Luigi,  Nisticò  Robert,  Zona  Cristina  and  Negri  Lucia…………17  

 • Pin1  and  Sirt1  gene  expression  changes   in  Alzheimer’s  Disease:   in  vitro  and   in  vivo  studies   -­‐  Lattanzio  Francesca,  

Carboni   Lucia,   Rimondini   Roberto,   Carretta   Donatella,   Mercatelli   Daniela,   Candeletti   Sanzio   and   Romualdi  Patrizia…………………………………………………………………………………………………………………………………………………………………..18  

 • Flavopiridol,   an     inhibitor   of   cyclin-­‐dependent   kinase   4,   reverses   cognitive   deficits   induced   by   β-­‐amyloid   1-­‐42   -­‐  

Leggio  Gian  Marco,  Pellitteri  Rosalia,  Copani  Agata,  Navarria  Andrea,  Catania  Maria  Vincenza,  Drago  Filippo,  Caraci  Filippo……………………………………………………………………………………………………………………………………………………………………19  

     

  5  

• Amyloid  beta  deregulates  astroglial  mGluR5-­‐mediated  Ca2+  signaling  via  calcineurin  and  NF-­‐kB  -­‐  Lim  Dmitry,  Grolla  Ambra,  Iyer  Anand,  Marcello  Elena,  Di  Luca  Monica,  Aronica  Eleonora  and  Genazzani  Armando  A……………………….20  

 • ADAM10   trafficking/endocytosis   in   dendritic   spines:   a   role   in   plasticity   and   Alzheimer’s   disease   pathogenesis   -­‐  

Musardo  Stefano,  Marcello  Elena,  Saraceno  Claudia,  Pelucchi  Silvia,  Gardoni  Fabrizio  and  Di  Luca,  Monica………….21    

• A  novel  binding  partner  of  ADAM10:  CAP2   -­‐  Pelucchi  Silvia,  Marcello  Elena,  Saraceno  Claudia,  Musardo  Stefano,  Gardoni  Fabrizio  and  Di  Luca  Monica…………………………………………………………………………………………………………………….22  

 • Pathogenetic   role   of   CSF   anti-­‐Aβ   autoantibodies   in   CAA-­‐related   inflammation   and   Alzheimer’s   disease:   a   novel  

biomarker  for  the  Amyloid  Related  Imaging  Abnormalities  for  amyloid-­‐modifying  therapies  -­‐  Piazza  F.,  Greenberg  S.M.,  Savoiardo  M.,  Giardinetti  M.,  Chiapparini  L.,  Raicher  I.,  Nitrini  R.,  Sakaguchi  H.,  Brioschi  M.,  Billo  G.,  Colombo  A.,  Lanzani  F.,  Piscosquito  G.,  Carriero  M.R.,  Giaccone  G.,  Tagliavini  F.,  Ferrarese  C.  and  DiFrancesco  J.C………………23  

 • Differential  deregulation  of  astrocytic  calcium  signaling  by  Amyloid  beta,  TNFalpha  and  LPS  -­‐  Ronco  Virginia,  Lim  

Dmitry  and  Genazzani  Armando  A…………………………………………………………………………………………………………………………24    

• SAP97  is  involved  in  PKC  activation  of  ADAM10  α-­‐secretase  -­‐  Saraceno  Claudia,  Marcello  Elena,  Musardo  Stefano,  Pelucchi  Silvia,  Gardoni  Fabrizio  and  Di  Luca  Monica……………………………………………………………………………………………..25  

 • Nilotinib  or  Dasatinib,  but  not  Imatinib,  might  reduce  plasma  beta-­‐amyloid  in  leukemia  patients:     implications  for  

Alzheimer’s  disease  -­‐  Tremolizzo  Lucio,  Conti  Elisa,  Zoia  Chiara  Paola,  Rizzo  Christian,  Gambacorti-­‐Passerini  Carlo,  Weksler  Marc  E.,  Weksler  Babette,  and  Ferrarese  Carlo………………………………………………………………………………………..26  

 • ERK   signalling   modulates   APP   metabolism,   tau   phosphorylation   and   EAAT1   processing   in   fibroblasts   from  

Alzheimer’s  patients  -­‐  Zoia  Chiara  Paola,  Conti  Elisa,  Costantino  Gisella,  Susani  Emanuela,  Isella  Valeria,  Tremolizzo  Lucio  and  Ferrarese  Carlo………………………………………………………………………………………………………………………………………27      

  6  

 Biliverdin  Reductase-­‐A:  a  Novel  Drug  Target  for  Atorvastatin  in  a  Dog  Preclinical  Model  of  Alzheimer  Disease.      Barone  Eugenio1,3,  Head  Elizabeth2  and  Butterfield  D.  Allan3.  1Brain  Mind  Institute,  School  of  Life  Sciences,  Swiss  Federal  Institute  of  Technology  (EPFL),  Station  15  CH-­‐1015  Lausanne,  Switzerland  

2Department   of   Molecular   and   Biomedical   Pharmacology   and   Sanders-­‐Brown   Center   on   Aging,  University  of  Kentucky,  Lexington,  KY,  USA  

3Department   of   Chemistry,   Center   of   Membrane   Sciences,   and   Sanders-­‐Brown   Center   on   Aging,  University  of  Kentucky,  Lexington,  KY  40506-­‐0055,  USA  

 

 

Biliverdin  reductase-­‐A  (BVR-­‐A)   is  a  pleiotropic  enzyme   involved   in  cellular  stress  responses.   It  not  only  transforms   biliverdin-­‐IX   alpha   into   the   antioxidant   bilirubin-­‐IX   alpha   but   through   its  serine/threonine/tyrosine  kinase  activity  is  able  to  modulate  cell  signaling  networks.  Interestingly,  BVR-­‐A  interacts  with  members  of  the  mitogen  activated  protein  kinase  family,  in  particular,  the  extracellular  signal-­‐regulated  kinases  1/2  (ERK1/2),  and  regulates  the  expression  of  oxidative-­‐stress   (OS)-­‐responsive  genes   such   as   heme-­‐oxygenase-­‐1   or   inducible   nitric   oxide   synthase.  We   previously   reported   BVR-­‐A’s  involvement   in   Alzheimer   disease   (AD)   and   amnestic   mild   cognitive   impairment   (MCI),   by   showing  phosphorylation   and   oxidative   differences   along   with   the   levels   and   activity   of   BVR-­‐A   in   brain   and  plasma  of  AD  or  MCI  subjects.    Statins,  a  class  of  hypolipidemic  drugs,  have  been  proposed  to  reduce  risk  of   incidence  of  AD.  In  this  study  we  evaluated  the  effect  of  atorvastatin  treatment  (80  mg/day  for  14.5  months)  on  OS  levels  and  BVR-­‐A  in  the  parietal  cortex,  cerebellum  and  liver  of  a  well  characterized  pre-­‐clinical   model   of   AD,   the   aged   beagles.   We   found   that   atorvastatin   significantly   i)   reduced   4-­‐hydroxy-­‐2-­‐nonenals   (HNE),   protein   carbonyls   (PC)   and   3-­‐nitrotyrosine   (3-­‐NT),   ii)   increased  GSH   levels,  and  iii)  increased  BVR-­‐A  protein  levels,  phosphorylation  and  activity  only  in  parietal  cortex.  Additionally,  significant   correlations   between   i)   decreased   levels   of   OS   markers   and   decrease   in   discrimination  learning  error   score   (DLES)   (reflecting   improved  cognition)  and   ii)  BVR-­‐A  and  decreased  OS   indices,  as  well   as   DLES   were   observed.   Furthermore   BVR-­‐A   up-­‐regulation   and   post-­‐translational   modifications  significantly  correlated  with  β-­‐secretase  protein  levels  in  the  brain,  suggesting  a  possible  role  for  BVR-­‐A  in   Aβ   formation.   These   observations   propose   a   novel   mechanism   of   action   for   atorvastatin   which,  through  the  induction  of  BVR-­‐A  post-­‐translational  modifications  may  contribute  to  the  neuroprotective  effects  of  this  enzyme,  thus  suggesting  a  potential  therapeutic  role  of  BVR-­‐A  in  AD.  

   

  7  

 CHF5074  restores  visual  memory  ability  and  rescue  synaptic  dysfunction  in  pre-­‐plaque  Tg2576  mice.      Beggiato  Sarah1,  Giuliani  Alessandro2,  Baldassarro  Vito  Antonio3,  Mangano  Chiara4,  Giardino  Luciana2,3,4,  Antonelli  Tiziana1,4,  Calzà  Laura2,3,4,  Ferraro  Luca4,5,  Imbimbo  Bruno  Pietro6.    1Dept.  of  Medical  Sciences,  University  of  Ferrara;  2Dept.  of  Veterinary  Medicine  and  Health  Science  and  3Technologies  Interdepartmental  Center  for  Industrial  Research  (HST-­‐ICIR),  University  of  Bologna;  4IRET-­‐ONLUS  Foundation,  Ozzano  Emilia,  Bologna;  5Dept.  of  Life  Sciences  and  Biotechnology  (SVEB),  University  of  Ferrara,  Italy  6Research  &  Development,  Chiesi  Farmaceutici,  Via  Palermo  26/A,  43100,  Parma.        Synaptic  dysfunction   is  an  early  event   in  Alzheimer's  disease   (AD)  and  occurs  before   the   formation  of  amyloid   plaques   and   neurofibrillary   tangles   [1].   In   particular,   the   appearance   of   cholinergic   neuritic  dystrophy,   i.e.   aberrant   fibers   and   fiber   swelling   are   widely   common   in   AD   [1].   It   has   been   recently  suggested   that   memory   impairment   in   plaque-­‐free   Tg2576   mice   may   be   due   to   cholinergic   synapse  dysfunction   rather   than   amyloid   plaque   deposition   [2].   Thus,   we   used   Tg2576   mice   to   compare   the  effects  of  CHF5074,  a  drug  which  attenuates  memory  deficit   in  AD  transgenic  mice   [3],  and  LY450139  (semagacestat)   on   in   vivo   novel   object   recognition   test   and  on   [3H]acetylcholine   and  GABA   release   in  pre-­‐plaque  (7  month-­‐old)  Tg2576  mice.  Vehicle-­‐treated   Tg2576  mice   displayed   an   impairment   of   recognition  memory   compared   to  wild-­‐type  animals.  This  impairment  was  recovered  in  transgenic  animals  acutely  treated  with  CHF5074  (30  mg/kg),  while   LY450139   (1-­‐10   mg/kg)   was   ineffective.   In   frontal   cortex   synaptosomes   from   vehicle-­‐treated  Tg2576  mice,  K+-­‐evoked  [3H]acetylcholine  release  was  lower  than  that  measured  in  wild-­‐type  mice.  This  reduction  was  absent  in  transgenic  animals  sub-­‐acutely  treated  with  CHF5074  (30  mg/kg  daily;  8  days),  while   it   was   slightly,   not   significantly,   amplified   by   LY450139   (3  mg/kg   daily;   8   days).   There  were   no  differences   between   the   groups   on   spontaneous   [3H]acetylcholine   release   as  well   as   on   spontaneous  and  K+-­‐evoked  GABA  release.  These  results  suggest  that  the  positive  effect  of  CHF5074  on  learning  and  memory  in  pre-­‐plaque  Tg2576  mice   is   associated  with   the   restore   of   K+-­‐evoked   acetylcholine   release   from   cortical   nerve   terminals,  which   is   decreased   in   vehicle-­‐treated   Tg2576   compared   to   wild-­‐type   animals.   Taken   together,   these  findings  suggest  CHF5074  as  a  possible  candidate  for  early  AD  therapeutic  regimens.      [1]Schliebs  and  Arendt,  2011,  Behav.  Brain  Res.  221,  555-­‐563.  [2]  Watanabe  et  al.,  2009,  Brain  Res.  1249,  222-­‐228.  [3]  Imbimbo  et  al.,  2010,  J.  Alzheimers  Dis.  20,  159-­‐73.            

  8  

 HMGB-­‐1  protein  and  β-­‐amyloid  oligomers  promote  neuronal  differentiation  of  adult  hippocampal  neural  progenitors  via  RAGE/NF-­‐κB  axis:  relevance  for  Alzheimer’s  Disease.      Bortolotto  Valeria,  Meneghini  Vasco,  Francese  Maria  Teresa,  Dellarole  Anna,  Carraro  Lorenzo,  Terzieva  Slavica  and  Grilli  Mariagrazia.    Department  of  Pharmaceutical  Sciences,  University  of  Piemonte  Orientale  Amedeo  Avogadro,  Novara  28100,  Italy        Dysregulated   hippocampal   neurogenesis   has   been   associated   with   neurodegenerative   disorders,  including  Alzheimer’s  disease  (AD),  in  which  it  may  potentially  represent  an  auto-­‐reparatory  mechanism  that  could  counteract  neuronal  loss  and  cognitive  impairment.  Although  literature  data  suggest  that  the  Receptor   for   Advanced   Glycation   End-­‐products   (RAGE)   plays   a   deleterious   role   in   AD,   recently   we  demonstrated   that   activation   of   the   RAGE/nuclear   factor-­‐κB   (NF-­‐κB)   axis   promotes   both   proliferation  and   neuronal   differentiation   of   adult   SVZ   neural   progenitor   cells   (NPC)   in   vitro.   Based   on   these  observations,   we   decided   to   investigate   the   role   of   the   RAGE/NF-­‐κB   axis   in   the  modulation   of   adult  hippocampal  neurogenesis  and  its  potential  relevance  in  AD  pathophysiology.  When  hippocampal  NPC  were   isolated   from   TgCRND8  mice,   we   demonstrated   that   (1)   their   neurogenic   potential   was   higher  compared  with  WT  NPC;  (2)  medium  conditioned  by  TgCRND8  NPC  promoted  neuronal  differentiation  of  WT  NPC;   and   (3)   the   proneurogenic   effect   of   TgCRND8-­‐conditioned  medium  was   counteracted   by  blockade  of  the  RAGE/NF-­‐κB  axis.  Furthermore,  we  showed  that  RAGE   ligands  such  as  β-­‐amyloid  1–42  oligomers,   but   not   monomers   and   fibrils,   and   the   alarmin   HMGB-­‐1   could   promote   neuronal  differentiation  of  NPC  via  activation  of  the  RAGE/NF-­‐κB  axis.  Altogether,  these  data  suggest  that,  in  AD  brain,  an  endogenous  proneurogenic  response  could  be  potentially  triggered  and  involve  signals  (Aβ1–42   oligomers   and   HMGB-­‐1)   and   pathways   (RAGE/NF-­‐κB   activation)   that   also   contribute   to  neuroinflammation/neurotoxicity.        

  9  

 JNK  mediated  Aβ  oligomers  synaptic  dysfunction.      Colombo  I.,  Sclip  A.,  Veglianese  P.,  Messa  M.,  Colombo  L.,  Salmona  M.  and  Borsello  T.    IRCCS  -­‐  Mario  Negri  Institute  for  Pharmacological  Research,  Milan,  Italy        

Alzheimer’s  disease   (AD)   is   a  progressive  neurodegenerative  disorder   that  begins  with  episodic   short-­‐term  memory   deficits   and   culminates  with   cognitive   impairment   and  memory   loss.   The   first   signs   of  memory  loss  are  due  to  synaptic  dysfunction,  which  result  from  the  toxicity  of  soluble  oligomeric  forms  of  beta  amyloid  (Aβ).  

The  molecular  mechanisms  by  which  soluble  Aβ  oligomers  are  responsible  for  synaptic  injury  still  remain  unclear  but  may  involve  mitogen-­‐activated  protein  kinases  (MAPKs).  Amongst  MAPKs,  c-­‐jun  N-­‐terminal  kinase   (JNK)  has  been  extensively   studied   for   its   role   in  AD  pathology.   To   investigate   the   intracellular  mechanisms  that  lead  to  AD  synaptopathy  we  set  up  an  new  in  vitro  model  of  synaptic  degeneration  by  treating   hippocampal   neurons   from   Brainbow   mice   with   sub-­‐toxic   concentrations   of   synthetic   Aβ  oligomers.  Following  Aβ  oligomers  application  we  reported  a  strong  activation  of  the  JNK  pathway  in  the  synaptic  compartment.  This  correlated  with   the   reduction  of  dendritic   spines  density,   the  decrease  of  postsynaptic  markers  (AMPAR  and  NMDAR  subunits,  PSD95  and  drebrin)  and  the  activation  of  caspase-­‐3  in   the   postsynaptic   compartment.   To   confirm   the   involvement   of   JNK   in   synaptic   degeneration  mechanisms   induced   by   Aβ   oligomers,   we   used   the   specific   cell   permeable   JNK   inhibitor   peptide,   D-­‐JNKI1.  Treatment  with  D-­‐JNKI1   reverted   the  synaptic  degeneration  by  preventing   the   loss  of  dendritic  spines   and   the   reduction   of   AMPAR   and  NMDAR   subunits,   PSD95   and   drebrin   from   the   postsynaptic  membrane.  Moreover   D-­‐JNKI1   treatment   prevented   caspase-­‐3   activation.   In   conclusion,   JNK   is   a   key  signalling  pathway  in  the  early  events  of  synaptic  degeneration  that  characterise  AD.          

  10  

 Role  of  donepezil  in  influencing  endogenous  immune  response  against  Abeta,  an  ex  vivo  and  in  vitro  study.      Conti  Elisa1,  Tremolizzo  Lucio1,  Santarone  Marta  Elena1,  Tironi  Marco1,  Zoia  Chiara  Paola1,  Appollonio  Ildebrando1  and  Ferrarese  Carlo1  

 1Dept.  of  Neurology,  San  Gerardo  Hospital  and  Department  of  Surgery  and  Interdisciplinary  Medicine,  University  of  Milano-­‐Bicocca,  Monza,  Italy        Donepezil   (DNPZ)   is   an   acetyl-­‐cholinesterase   inhibitor   (AChEI)   approved   for   the   treatment   of  mild-­‐to  moderate  Alzheimer’s  disease  (AD).  DNPZ  efficacy  can  be  attributed  to  an  improvement  of  acetylcholine  (ACh)  transmission  as  well  as  to  further  mechanisms,  among  which  the  capacity  to  reduce  toxic  Abeta  fibrils   and   a   modulation   of   the   immune   response   have   been   hypothesized.   In   a   previous   study   we  demonstrated  that  AD  patients  treated  with  AChEI  showed  a  specific  increase  of  plasma  anti-­‐Abeta1-­‐42  antibodies  when  compared   to  untreated  patients.   In   the  present   study  we  evaluated   the   influence  of  DNPZ   in   favouring   a   Th2   phenotype,   involved   in   modulating   the   immune   humoral   response.   We  hypothesized   that   this  mechanism  might   be  mediated   by   the   a7-­‐nicotinic   ACh   receptor   expressed   in  lymphocytes.  60  patients  with  mild  or  moderate  AD,  either  treated  (n=22)  or  not  (n=38)  with  DNPZ,  and  30  controls  were  enrolled.  AD  DNPZ+  showed  significantly  higher  plasma  levels  of  anti-­‐Abeta  antibodies  than  DNPZ-­‐  (+40%)   and   lower   levels   of   Abeta   1-­‐42   than   controls   (-­‐50%).   In   a   subgroup   of   subjects,   GATA-­‐3,   a  transcription   factor   involved   in   Th2   differentiation,   and   α7nAChR   expression   was   evaluated.   No  differences  were  found  in  GATA-­‐3  mRNA  in  AD  DNPZ+  when  compared  to  DNPZ-­‐  and  controls.  On  the  opposite,  we   found  by   chromatine   immunoprecipitation  assay,   a   significant   three-­‐fold   increase  of   the  association   of   GATA-­‐3   with   the   IL-­‐5-­‐promoter   in   DNPZ+   patients,   which   also   showed   a   significant  twofold   increase  in  α7-­‐nAChR  mRNA,  with  respect  to  DNPZ-­‐  ones.   In  vitro  analyses  demonstrated  that  the   capacity   of  DNPZ   to  modulate  GATA-­‐3   expression   is  mediated   by  α7nAChR,   since  MLA,   a   specific  antagonist,  prevents  it.  Further  studies  are  needed  to  better  understand  the  role  of  DNPZ  in  modulating  the  immune  response  against  Abeta,  possibly  ameliorating  therapeutic  strategies  for  AD.      

  11  

 Acety-­‐L-­‐carnitine   is   a   potent   positive  modulator   of   adult   hippocampal   neurogenesis   in   vitro   and   in  vivo.      Cuccurazzu   Bruna1,2,   Bortolotto   Valeria1,2,   Valente   Maria   Maddalena   1,2,   Ubezio   Federica1,2,   Canonico  Pier  Luigi  2    and  Grilli  Mariagrazia  1,2.    1Laboratory   of   Neuroplasticity   and   Pain,   2Dept.   of   Pharmaceutical   Sciences,   University   of   Piemonte  Orientale  “A.  Avogadro”,  Novara,  Italy  28100        Endogenous  acetyl-­‐L-­‐Carnitine  (ALC),  aside  from  its  role  in  cellular  bioenergetics,  modulates  the  activity  of   neurotrophic   factors,   hormones,   neurotransmitters   in   nervous   tissues.   Moreover   exogenously  administered  ALC  can  readily  pass  the  blood-­‐brain-­‐barrier  and  is  neuroprotective  at  supraphysiological  concentrations   (Jones   et   al,   2010).   In   humans   beneficial   effects   of   ALC   were   suggested   in   Alzheimer  disease   and   age-­‐related   cognitive   impairment   (Montgomery   et   al,   2003).   Here   we   report   a   novel  pharmacological  activity  of  ALC,  namely  its  potent  proneurogenic  effects  in  vitro  and  in  vivo.  Specifically,  ALC   dramatically   promoted,   in   a   concentration   dependent   manner,   neuronal   differentiation   of   adult  hippocampal  neural  progenitors  and  these  effects  were  independent  of  its  neuroprotective  activity.  ALC  proneurogenic  effects  appeared  to  be  mediated  by  activation  of  the  NF-­‐κB  pathway,  and  in  particular  by  its   ability   to   promote   p65   acetylation   and   subsequent   NF-­‐κB-­‐mediated   upregulation   of  metabotropic  glutamate   receptor   2   (mGlu2)   receptor   expression.  Moreover   in   vivo   21   days   of   ALC   treatment   (100  mg/kg,   s.c.)   significantly   increased   hippocampal   neurogenesis   in   adult   mice.   Since   ALC   is   very   well-­‐tolerated  in  humans,  these  data  suggest  the  importance  of  further  investigating  the  effects  of  ALC  in  AD  animal  models  where  proneurogenic  drugs  have  shown  ability  to  attenuate  cognitive  impairment.        

  12  

 Neuroprotection   by   association   of   palmitoylethanolamide   with   luteolin   (Ġlìalia®)   in   experimental  Alzheimer’s  disease  models:    the  control  of  neuroinflammation.      Esposito  Emanuela,  Paterniti  Irene,  Campolo  Michela  and  Cuzzocrea  Salvatore    Department  of  Biological  and  Environmental  Sciences,  University  of  Messina        Activation  of  glial  cells  and  the  consequent  neuroinflammatory  response  is  increasingly  recognized  as  a  prominent   neuropathological   feature   of  Alzheimer’s   disease   (AD)   (Scuderi   et   al.,   2012).   Targeting   the  signaling   pathways   in   glial   cells   responsible   for   neuroinflammation   represents   a   promising   new  therapeutic   approach   designed   for   AD.   We   choose   to   use   in   our   studies   the   endocannabinoid  palmitoylethanolamide   (PEA)  with  well   known   anti-­‐inflammatory   effects,   and   the   antioxidant   luteolin  (Lut),  to  counteract  the  neuroinflammation  of  AD.  In  the  present  study  we  assessed  the  neuroprotective  effect   of     association   of   PEA   with   Lut   (PEA-­‐Lut,   Ġlìalia®),   a   compound   obtained   by   a   co-­‐ultramicronization  process,  and  its  relevance  to  AD  sintomatology  in  in  vitro  and  in  ex  vivo  organotypic  model  of  AD.  Human  neuroblastoma  cell   line  SH-­‐SY5Y,   that  was  differentiated  with   retinoic  Acid   (100  nM),  and  rat  hippocampal  organotypic  were  pre-­‐treated  with  PEA-­‐Lut  (at  three  different  concentration  0.1-­‐1-­‐10  μM)  for  2h.  The  damage  was  induced  by  Aβ1-­‐42  (1µM)  for  24h;  after  damage  we  performed  cell  death  assay  and  western  blot  analysis.  We  will  study  the  effects  of  PEA-­‐Lut  on  the  microglial  activation,  GFAP-­‐positive  astrocytes,  nNOS  positive  cells  and  MMPs  expression  induced  by  Aβ1-­‐42.  Moreover,  mRNA  and   protein   expression   will   be   carried   out   for   GFAP,   CD11b,   TNFα,   IL1β,   iNOS,   and   COX-­‐2.   Our   data  indicate  that  PEA-­‐Lut  compound  is  able  to  blunt  A  Aβ1-­‐42  -­‐induced  neurotoxicity  and  to  exert  a  marked  protective  effect  on  glial  cells.  These  findings  highlight  new  pharmacological  properties  of  PEA-­‐Lut  and  suggest  that  this  compound  may  provide  an  effective  strategy  for  AD.    Scuderi  C  et  al.  J  Neuroinflammation.  2012  Mar  9;9:49.      

  13  

 SUMOylation/deSUMOylation:  a  balance  to  be  re-­‐equilibrated.      Feligioni  Marco1  and  Nisticò  Robert2,3  

 1.  EBRI  ‘Rita  Levi-­‐Montalcini’  Foundation,  Synaptic  Plasticity  laboratory,  Rome  2.  Sapienza  University  of  Rome,  Dept  of  Physiology  and  Pharmacology,  Rome  3.  IRCCS  S.Lucia  Foundation,  Rome        Oxidative  stress  is  a  central  feature  of  the  pathogenesis  of  Alzheimer  disease  and  protein  SUMOylation  seems  to  play  a  key  role  in  this  process  [1].  AβPP,  Tau  and  related  proteins  (JNKs,  BACE1,  GSK3-­‐β)  can  be  conjugated   by   SUMO   [2,3],   and   this   event   leads   to   AβPP   and   Tau   misprocessing.   Moreover   protein  SUMOylation   is   also   important   in   controlling   synaptic   plasticity   in   normal   and  pathological   conditions  [4,5].   Thus,   we   hypothesize   that   unbalance   in   SUMO/deSUMOylation   equilibrium   could   alter   specific  intracellular  signalling  pathways  promoting  AD  pathogenesis.  Preliminary  data   show  that  over-­‐expressed  catalytic  domain  of  deSUMOylation  enzyme   (SENP1-­‐CD)   in  H4swe  cells   (in  vitro  model  of  AD)   reduced  AβPP  phosphorylation  while  SUMO-­‐1  over-­‐expression  had  the   opposite   effect,   suggesting   that   deSUMOylation   inhibits   phosphorylation   of   AβPP.   Other  experiments  showed  an  increment  of  protein  SUMOylation  in  the  hippocampus  of  3  month-­‐old  (disease  onset)   AD  mice   (Tg2576)   vs   control.   This   led   us   to   hypothesize   that   in   the   early   stages   of   disease   an  increased  SUMOylation  state  occurs.  We  then  produced  a  cell  permeable  peptide  (TAT-­‐SENP1-­‐CD),  exploiting  TAT  cargo  strategy,   to  reduce  protein   SUMOylation.   The   catalytic   gene   of   deSUMOylation   enzyme   SENP1  was   inserted   into   a   pTAT  vector.  Remarkably,  TAT-­‐SENP1-­‐CD  was  able  to  reverse  LTP   impairment   in  12  month-­‐old  Tg2576  mice,  further   suggesting   that   SUMOylation   plays   a   critical   role   in   synaptic   plasticity   [5]   and   its  modulation  could  be  relevant  to  counteract  synaptic  dysfunction  underlying  AD.      References  [1]  PloS  one,  vol.  6,  no.  12,  Jan.  2011.  [2]  Experimental  Neurol,  no.  2,  pp.  322–5,  Jun.  2010.  [3]  J  of  Neurosci,  no.  28,  pp.  9078–89,  Jul.  2009.  [4]  Curr  Opin  Neurobiol.  2012  Jun;22:480-­‐7.  [5]  Eur  J  Neurosci,pp.1348–56,  Apr.  2009.      

  14  

 β-­‐amyloid   differently   regulates   the   function   of   nAChRs   which   stimulate   the   release   of   glutamate,  aspartate,  glycine  and  GABA  in  rat  hippocampus.      Grilli   Massimo1,   Zappettini   Stefania1,   Mura   Elisa2,   Preda   Stefania2,   Salamone   Alessia1,   Olivero  Guiendalina1,  Govoni  Stefano2      and  Marchi  Mario1.      Department  of  Pharmacy,  School  of  Medicine,  University  of  Genoa,   Italy1;  Department  of  Experimental  and  Applied  Pharmacology,  Centre  of  Excellence  in  Applied  Biology,  University  of  Pavia,  Italy2        Early  stages  of  Alzheimer  disease  are  associated  with  learning  impairments  and  cognitive  decline  which  have  been  linked  to  altered  transmission  at  excitatory  synapses  in  the  hippocampus.  This  presentation  will   describe   some   new   neuromodulatory   effects   of   beta   amyloid   Aβ1-­‐40   on   the   nicotinic-­‐evoked  aminoacid   release   in   rat   hippocampus.   The   methodological   approach   included   in   vivo   microdialysis  technique   and   in   vitro   release   from   superfused   synaptosomes   and   gliosomes   (releasing   particles  prepared  from  astrocytes).  Nicotine  and  some  selective  agonists  greatly  enhanced  the  in  vivo  release  of  endogenous  GLU,  ASP,  GLY  and  GABA  indicating  the  involvement  of  both  α7  and  α4β2  nAChR  subtypes.  The  stimulatory  effect  produced  by  activation  of  α7  nAChRs  was  predominant  over   that  of  α4β2.  The    Aβ1-­‐40     which     was   ineffective   on   basal   aminoacid   release   significantly     inhibited   GLU,   ASP   and   GLY    release    evoked  by  nicotine  but  produced  a  biphasic  effect  (inhibitory  and  stimulatory)  on  the  nicotine-­‐evoked    in  vivo  GABA  release.  The  in  vitro  results  confirm  the  inhibitory  effect  of  Aβ1-­‐40  on  the  α7  and  α4β2   nAChR   subtypes   modulating   aminoacids   release   present   on   synaptosomes   and   gliosomes.    However,  at  picomolar  concentration,  Aβ1-­‐40  increased  selectively  the  α7-­‐  but  not  the  α4β2  -­‐mediated  release  of  ASP  and  GLU  and  not  that  of  GABA  and  GLY.  The  apparent  discrepancy  between  in  vivo  and  in  vitro   results  may  be  due   to   the   interaction  of  hierarchically  organized   synapses   that  occurs   in  vivo.   In  conclusion  Aβ  1-­‐40  disrupted  the  cholinergic  control  of  GLU,  ASP,  GLY  and  GABA  release  modulating  in  a  different  manner  the  functional  responses  of  α7  and  α4β2  nAChRs.        

  15  

 Amyloid-­‐b   and  Alzheimer´s   disease   type  pathology  differentially   affects   calcium   signalling   toolkit   in  astrocytes  from  different  brain  regions.        Grolla   Ambra   A   1,2,   Sim   Joan   A   2,   Lim   Dmitry   1,   Rodriguez   Jose   Julio   3   ,   Genazzani   Armando   A   1   and  Verkhratsky  Alexei  2        1Dipartimento   di   Scienze   del   Farmaco,   Università   degli   Studi   del   Piemonte   Orientale   “Amedeo  Avogadro”,  Novara,  28100,  Italy;    2  Faculty  of  Life  Sciences,  University  of  Manchester,  Manchester  M13  9PL,  UK;    3,   IKERBASQUE,   Basque   Foundation   for   Science,   48011,   Bilbao,   and   Department   of   Neurosciences,  University  of  the  Basque  Country  UPV/EHU  &  CIBERNED,  48940,  Leioa,  Spain        Entorhinal-­‐hippocampal   circuit   is   severely   affected   in  Azheimer’s   disease   (AD).  Here,  we  demonstrate  that  amyloid-­‐b  (Ab)  differentially  affects  primary  cultured  astrocytes  derived  from  the  entorhinal  cortex  (EC)  and   from  the  hippocampus  of  non-­‐transgenic   controls  and  3xTg-­‐AD   transgenic  mice.  Exposure   to  100  nM  of  Ab  resulted  in  increased  expression  of  the  metabotropic  glutamate  receptor  type  5  (mGluR5)  and  its  downstream  InsP3  receptor  type  1  (InsP3R1)  in  hippocampal  but  not  in  EC  astrocytes.  Amplitudes  of  Ca2+   responses   to  an  mGluR5  agonist,  DHPG,  and   to  ATP,  another  metabotropic  agonist  coupled   to  InsP3Rs,  were  significantly  increased  in  Ab  treated  hippocampal  but  not  in  EC  astrocytes.  Previously  we  demonstrated  that  senile  plaque  formation  in  3xTg-­‐AD  mice  triggers  astrogliosis  in  hippocampal  but  not  in  EC  astrocytes.  Different  sensitivities  of  Ca2+  signalling  toolkit  of  EC  vs.  hippocampal  astrocytes  to  Ab  may  account  for  the  lack  of  astrogliosis   in  the  EC,  which  in  turn  can  explain  higher  vulnerability  of  this  region  to  AD.              

  16  

 Disease  modifying  effects  of  the  polyphenol  Oleuropein  Aglycone  on  Alzheimer’s  Disease:  a  Preclinical  Study.        Grossi  Cristina1,  Rigacci  Stefania2,  Ed  Dami  Teresa1,  Luccarini   Ilaria1,  Stefani  Massimo  2,3  and  Casamenti  Fiorella  1,3  

 1  Department  of  Neuroscience,  Psychology,  Drug  Research  and  Child  Health,  Division  of  Pharmacology  and  Toxicology,  University  of  Florence,  50139  Florence,  Italy.  2  Department  of  Experimental  and  Clinical  Biomedical  Sciences,  University  of  Florence,  50134  Florence,  Italy.  3  Research  Centre  on  the  Molecular  Basis  of  Neurodegeneration,  University  of  Florence,  50134    Florence,  Italy        Mounting  evidence  supports  the  beneficial  effects  of  the  Mediterranean  diet  in  preventing  age-­‐related  dysfunctions,  cancer,  neurodegenerative  diseases  and  in  attenuating  Alzheimer’s  disease-­‐like  pathology  and   cognitive   deterioration.   Focusing   dietary   regimens   associated  with   a   reduced   risk   of   Alzheimer’s  disease   in   the   aged   population   can   be   useful   to   find   molecules   exploitable   for   Alzheimer’s   disease  prevention   and   therapy.   The   effects   of  dietary   supplementation   of   oleuropein   aglycone   (50  mg/kg   of  diet),  the  main  polyphenol  in  extra  virgin  olive  oil,  were  investigated  on  cognitive  functions,  Aß  plaque  formation/disassembly   and   associated   neuroinflammation   in   TgCRND8  mice   of   3   (young),   6   (middle-­‐aged)   and   12   (old)   months.   Oleuropein   aglycone   administered   young-­‐   and   middle-­‐aged   Tg   mice   got  significantly  better  scores  in  the  step  down  test  and  novel  object  recognition  test  and  displayed  a  better  general  condition  as  compared  to  their  littermate  controls.  Oleuropein  aglycone  administration  robustly  prevented  Aß  deposition   in  TgCRND8  mice  as  demonstrated  by   the   significant   reduction     (P<0.001)   in    Aß  plaque  load  within  the  cortex  and  hippocampus    and  markedly  reduced  the  presence  of  dense-­‐cored  Aß   plaques   in   6-­‐   and   12-­‐month-­‐old   TgCRND8   mice.   Oleuropein   aglycone   administration   induced  substantial  differences   in  plaques  morphology,  reduced  Aß  deposits  compactness,  oligomerization  and  pyroglutamilation,  astrocyte  reaction  and  determined  microglia  migration  to  plaques  for  phagocytosis.  Moreover     oleuropein   aglycone   enhanced   the   autophagic   flux   as   shown   by   an   astonishingly   intense  elevation   of   the   autophagic   and   lysosomal  markers   in   TgCRND8  mice   at   all   ages.   Overall,   our   results  provide   a   strong   evidence   of   oleuropein   aglycone   neuroprotective,   beneficial   and   therapeutic   effects  and   indicate   that   dietary   supplementation   with   oleuropein   aglycone   may   be   prophylactic   for  Alzheimer’s   disease   or,   at   least,   suitable   to   delay   the   occurrence   and   to   reduce   the   severity   of   its  symptoms.  Supported  by  Salute  2009  Regione  Toscana,  Ente  CRF  2010-­‐2011,  PRIN  2008.      

  17  

 Bv8/prokineticin  2  is  a  potential  mediator  of  Alzheimer’s  disease.      Lattanzi  Roberta1,  Severini  Cinzia2,  Ciotti  Maria  Teresa2,  Petrocchi  Pamela2,  Marconi  Veronica1,  Giancotti  Luigi1,  Nisticò  Robert1,  Zona  Cristina3  and  Negri  Lucia2  

 1Department   of   Human   Physiology   and   Pharmacology   "Vittorio   Erspamer",   University   of   Roma   "La  Sapienza",  P.za  A.  Moro  5,  00185  Roma,  2Institute  of  Cell  Biology  and  Neurobiology,  CNR,  Via  del  Fosso  di  Fiorano,   64,   00143,   Roma,   3Department   of   Neuroscience,   University   of   Rome   "Tor   Vergata",   Via  Montpellier,  1,  00133  Rome.        Prokineticin  2  (PK2)  is  a  bioactive  peptide  initially  discovered  as  a  regulator  of  gastrointestinal  motility.    Multiple  biological   roles   for  PK2  have  been  discovered,   including  circadian  rhythms,  angiogenesis,  and  neurogenesis.  Recently,  PK2  has  been  identified  as  a  deleterious  mediator  for  cerebral  ischemic  injury.  Indeed,   PK2   can   be   activated   by   pathological   stimuli   such   as   hypoxia-­‐ischemia   and   excitotoxic  glutamate.  Aim  of  the  present  study  was  to  evaluate  the  PK2  potential  involvement  in  amyloid  beta  (Ab)  neurotoxicity,  the  characteristic  Alzheimer’s  disease  (AD)  insult.    Using  primary  cortical  cultures,  we  found  that  PK2  mRNA  is  up-­‐regulated  by  Ab  peptide,  suggesting   its  potential  involvement  in  AD.  We   therefore   characterized,   by   immunofluorescence,   the   presence   of   both   PK1   and   PK2   receptors   in  cortical   neurons.   In   view   of   their   presence,   we   tested   the   neuroprotective   activity   of   a   PK   receptor  antagonist  (PC-­‐1)  against  neuronal  death  induced  by  Ab.  We  found  that  PC-­‐1  dose-­‐dependently  protects  cortical   neurons   against   both   Ab25-­‐35-­‐   and   Ab1-­‐42-­‐induced   neurotoxicity,   as   revealed   by   live/dead   cell  assay  and  Hoechst   staining.   Electrophysiological   experiments   showed   that   the  Ab-­‐induced   increase  of  kainate   current   amplitude   was   reversed   by   PC-­‐1   treatment.  Moreover,   PC-­‐1   completely   rescued   LTP  impairment   in  hippocampal   slices   from  6  month-­‐old  Tg2576  AD  mice  without  affecting  basal   synaptic  transmission  and  paired  pulse-­‐facilitation  paradigms.    These   results   indicate   that  PK2  plays   a   role   in  Ab-­‐mediated  neuronal  death  and   that  PK2   receptor  antagonists  may  represent  a  new  approach  in  the  understanding  and  treatment  of  AD.        

  18  

 Pin1  and  Sirt1  gene  expression  changes  in  Alzheimer’s  Disease:  in  vitro  and  in  vivo  studies.      Lattanzio   Francesca,   Carboni   Lucia,   Rimondini   Roberto,   Carretta   Donatella,   Mercatelli   Daniela,  Candeletti  Sanzio  and  Romualdi  Patrizia    Department  of  Pharmacy  and  Biotechnology  (FaBiT),  University  of  Bologna,  Italy        Increasing  evidence  strongly  suggests  that  deregulation  of  sirtuin  1  (Sirt1)  and  peptidyl-­‐prolyl  cis-­‐trans  isomerase  (Pin1)  is  involved  in  the  early  physiopathological  stages  of  Alzheimer’s  disease  (AD).  Sirt1  is  a  deacetylase  enzyme  that  plays  an  important  role  in  the  synaptic  plasticity  and  seems  to  be  involved  in  the  reduction  of  amyloid-­‐beta  (Aβ)  deposition  and  tau  phosphorylation.  Pin1  isomerizes  phosphorylated  serine/threonine-­‐proline   motifs   of   several   proteins   regulating   their   conformational   changes   and  transductional   pathways.   Pin1   seems   to   prevent   the   aberrant   conformation   of   tau   protein,   reducing  neurofibrillary  tangles  formation,  and  also  regulates  amyloid  precursor  protein  (APP)  conformation  and  processing,   leading   to   reduced  amyloid-­‐beta  deposition.  Sirt1  and  Pin1  expression   inversely  correlates  with   neurodegeneration   in   AD   brains,   suggesting   their   protective   roles   against   pathological  modifications  of  APP  and  tau  proteins;  however  the  mechanism  is  not  completely  elucidated.  The  aim  of  the  present  study  was  to  evaluate  the  effect  of  the  risk  factors  apolipoprotein  E4  (apoE4)  genotype  and  Aβ  peptides  on  Pin1  and  Sirt1  gene  expression.    We   used   18   months   old   apoE3   and   apoE4   human   targeted   replacement   mice   and   SH-­‐SY5Y  neuroblastoma   cells   exposed   to   amyloid-­‐beta   peptide.   Sirt1   and   Pin1   gene   expression   levels   were  measured  by  Real  Time  PCR  assay.    The   in   vivo   results   showed   that   Sirt1   gene   expression   was   decreased   in   frontal   cortex;   Pin1   gene  expression   levels  were  significantly  decreased   in   the  entorhinal  and  parietal  cortices,  and   increased   in  the  hippocampus  of  apoE4  mice  as  compared  to  apoE3  controls.  In  SH-­‐SY5Y  cells  exposed  to  25  μM  Aβ  for  5  and  24  h,  Sirt1  was  decreased.  Pin1  gene  expression  was  also  decreased  in  cells  exposed  to  Aβ  for  24  h  No  changes  were  detected  at  48h.  The   present   data   suggest   that   Sirt1   and   Pin1   gene   expression   are  modulated   by   apoE4   and   amyloid-­‐beta.  Thus,  Sirt1  and  Pin1  might  represent  potential  diagnostic  and  therapeutic  targets  in  AD.        

  19  

 Flavopiridol,   an   inhibitor   of   cyclin-­‐dependent   kinase   4,   reverses   cognitive   deficits   induced   by   β-­‐amyloid  1-­‐42.      Leggio  Gian  Marco1  ,  Pellitteri  Rosalia2  ,  Copani  Agata3,  Navarria  Andrea1,  Catania  Maria  Vincenza2  ,  Drago  Filippo1,  Caraci  Filippo1,4    1Department   of   Clinical   and   Molecular   Biomedicine,   Section   of   Pharmacology   and   Biochemistry,  University  of  Catania,  Catania,  Italy;  2Istituto  di  Scienze  Neurologiche  CNR,  Catania,  Italy;  3Department  of  Drug  Sciences,  University  of  Catania,  Catania,   Italy;   4Department  of  Educational  Sciences,  University  of  Catania,  Catania,  Italy.        Activation   of   cell   cycle   is   considered   an   early   event   in   the   pathogenesis   of   Alzheimer’s   disease   (AD).  Expression   of   cell   cycle   proteins   and   replicative   DNA   synthesis   has   been   observed   in   neuronal  populations   fated  to  degenerate   in   the  AD  brain.  The  ectopic   re-­‐activation  of  cell   cycle   in  AD  neurons  could   lead   to   synaptic   failure   and   ensuing   cognitive   deficits   that   precede   frank   neuronal   death   in  AD  brain.   In  cultured  neurons,   synthetic  beta-­‐amyloid  peptide   (Aß)   reproduces   the  neuronal  cell   cycle   re-­‐entry   observed   in     transgenic   animals   and   in   the   AD   brain.   Cyclin-­‐dependent   kinase   (CDK)   inhibitors,  such  as  flavopiridol,  are  protective  in  vitro  against  Aß-­‐induced  toxicity,  but  their  effects  in  animal  models  of  AD  are  unknown.    We   examined   the   effect   of   flavopiridol   on   memory   retention   loss   in   CD1   mice   injected  intracerebroventricularly   (I.C.V.)   with   Aß   1-­‐42   (400   pmol/mouse).   Three   days   after   Aß   injection,  flavopiridol   (0.5,   1   and   3   mg/kg)   was   administered   intraperitoneally   (i.p.)   to   animals   for   11   days.  Flavopiridol   (0.5   and   1   mg/kg)   reversed   in   the   passive   avoidance   test   the   loss   of   memory   retention  induced  by  Aß1-­‐42.  Using  immuno-­‐histochemical  procedures  we  found  a  strong  induction  of  cyclin  A2  in  NeuN   positive   cells   of   cerebral   cortex,   hippocampus   and   amygdala   of   Aβ-­‐injected  mice   compared   to  vehicle-­‐treated   animals.   Interestingly,   flavopiridol   at   the   lowest   dose   of   0.5   mg/kg   prevented   the  occurrence   of   ectopic   cell-­‐cycle   events   in   cerebral   cortex,   hippocampus   and   amygdala.These   data  suggest  for  the  first  time  that  drugs  that  inhibit  cell  cycle  activation,  such  as  flavopiridol,  might  prevent  cognitive  deficits  in  AD.      

  20  

 Amyloid  beta  deregulates  astroglial  mGluR5-­‐mediated  Ca2+  signaling  via  calcineurin  and  NF-­‐kB.      Lim   Dmitry1,   Grolla   Ambra1,   Iyer   Anand2,   Marcello   Elena3,   Di   Luca   Monica3,   Aronica   Eleonora2   and  Genazzani  Armando  A1    1Dept  Pharmaceutical  Sciences,  Amedeo  Avogadro  Univ,    Novara,  Italy.  2Dept  (Neuro)Pathology,  Univ  of  Amsterdam,  Amsterdam,  The  Netherlands.  3Dept  Pharmacological  Sciences,  Univ  of  Milan,  Milan,  Italy.        Deregulation   of   calcium   signaling   in   astrocytes   accompany   neuronal   dysfunction   in   several  neurodegenerative   diseases   including   Alzheimer’s   disease   (AD),   although   mechanisms   of   such  deregulation   are   not   understood.   Here   we   demonstrate   the   ability   of   astrocytes   to   participate   in  generation   of   toxic   amyloid   beta   (Aβ)   peptide   and   dissect   a   cascade   of   signaling   events   by  which   Aβ  deregulates  astroglial  Ca2+  signaling.  Using  a  cell  permeable  peptide,  termed  Tat-­‐Pro,  which  disrupts  the  complex   between   SAP97   and   the   α-­‐secretase   ADAM10,   thereby   shifting   the   APP   processing   towards  amyloidogenic   process,   we   report   that   Tat-­‐Pro-­‐treated   astrocytes   secrete   increased   amount   of   both  Aβ42   and   Aβ40.   Moreover,   our   data   demonstrate   that   hippocampal   astrocytes   express   all   the  components   for   the   amyloidogenic   and   non-­‐amyloidogenic   processing   of   APP.   Tat-­‐Pro,   as   well   as  exogenous  Aβ,  deregulates  Ca2+  homeostasis   through   increased  expression  of  key  components  of   the  astroglial  Ca2+  signaling,  mGluR5  and  IP3  receptors.  Our  data  demonstrate  that:  100  nM  Aβ  leads  to  an  increase   in   cytosolic   calcium   that   results   in   activation   of   calcineurin   (CaN)   that   indirectly   activates  transcription  factor  NF-­‐kB,  that  was  followed  by  increase  of  mGluR5  and  IP3R2  mRNA  after  24  hours  of  treatment.   The   transcriptional   effects  were  blocked  by   inhibitors  of  CaN  and  NF-­‐kB.   Furthermore,  we  show  that  Aβ  treatment  of  glial  cells   leads  to  de-­‐phosphorylation  of  Bcl10  and  an  increased  CaN-­‐Bcl10  interaction.   In   support   of   these   results,   mGluR5   staining   is   augmented   in   hippocampal   astrocytes   of  Alzheimer’s  disease  patients   in  proximity  of  Aβ  plaques  and  co-­‐localizes  with  nuclear  accumulation  of  the  p65  and   increased   staining  of  CaNAα.  Taken   together,  our   results   suggest   that   astroglia  might  be  active  players  in  Aβ  production  and  indicate  that  nanomolar  [Aβ]  deregulates  Ca2+  homeostasis  via  CaN  and  its  downstream  target  NF-­‐kB,  possibly  via  the  cross-­‐talk  of  Bcl10  in  hippocampal  astrocytes.      

  21  

 ADAM10   trafficking/endocytosis   in   dendritic   spines:   a   role   in   plasticity   and   Alzheimer’s   disease  pathogenesis.      Musardo  Stefano1;  Marcello  Elena1;  Saraceno  Claudia1;  Pelucchi  Silvia1;  Gardoni  Fabrizio1  and  Di  Luca,  Monica1    

1Department  of  Pharmacological  and  Biomolecular  Sciences,  University  of  Milan,  via  Balzaretti  9,  20133  Milan,  Italy        Alzheimer’s  disease  (AD)  is  the  most  common  neurodegenerative  disorder  characterized  by  progressive  loss  of  synapses  and  neurons  and  accumulation  of   insoluble  deposits  of  amyloid  beta-­‐peptide  (Abeta).  Although  AD  is  emerging  as  the  most  prevalent  and  socially  disruptive  illness  of  aging  populations,  it  is  currently  incurable.  Abeta   derives   from   the   amyloid   precursor   protein   (APP),   which   can   follow   2   mutually   exclusive  pathways   in   the   cell.   The   amyloidogenic   pathway   involves   BACE   and   gamma   secretase   activities   and  leads  to  Abeta  formation.  On  the  other  hand,  the  main  protagonist  of  the  non-­‐amyloidogenic  pathway  is  ADAM10,   a   disintegrin   and  metalloproteinase   10,  which   cleaves   APP   in   the   domain   corresponding   to  Abeta,  thus  precluding  Abeta  production.  ADAM10   cleaves   its   substrates   when   correctly   localized   at   the   plasma   membrane,   therefore   the  mechanisms   regulating   its   trafficking  can  affect   its  activity.   Since   the  modulation  of  ADAM10  synaptic  localization  through  ADAM10  membrane   insertion/removal  could  constitute  an   innovative  therapeutic  strategy  to  finely  tune  its  shedding  activity,  we  have  investigated  the  mechanisms  underlying  ADAM10  endocytosis.   We   show   that   ADAM10   removal   from   the   plasma   membrane   is   mediated   by   clathrin-­‐dependent  endocytosis  and  we  describe  the  clathrin  adaptor  AP2,  a  heterotetrameric  assembly  which  initiates   the   endocytosis   process,   as   new   interacting   partner   of   ADAM10   C-­‐terminal   domain.   In  particular,  we   identify  an  atypical  binding  motif   for  AP2  complex   in  ADAM10  cytoplasmic  tail,  which   is  relevant   for   ADAM10   endocytosis   and   the  modulation   of   its   plasma  membrane   levels.  Moreover,  we  describe  a  pathological  alteration  of  ADAM10/AP2  association  in  AD  and  a  physiological  role  in  activity-­‐dependent   synaptic   plasticity.   We   demonstrate   that   long-­‐term   potentiation   induces   ADAM10  endocytosis,   through  AP2   association,   and   decreases   surface   ADAM10   levels   and   activity,  while   long-­‐term  depression  promotes  ADAM10  synaptic  membrane  insertion  and  stimulates  its  activity.  Finally   we   have   designed   different   cell   permeable   peptides   able   to   interfere   with   ADAM10/AP2  association  and,  thereby,  to  reduce  ADAM10  endocytosis.      

  22  

 

A  novel  binding  partner  of  ADAM10:  CAP2.      Pelucchi   Silvia,   Marcello   Elena,   Saraceno   Claudia,   Musardo   Stefano,   Gardoni   Fabrizio   and   Di   Luca  Monica.    

Department  of  Pharmacological  and  Biomolecular  Sciences  -­‐  University  of  Milan  Via  Balzaretti  9,  20133  Milan,  Italy        Alzheimer   disease   (AD)   is   a   progressive   and   neurodegenerative   disorder   characterized   by   increased  levels  of  amyloid  β-­‐peptides  (Aβ)  and  their  deposition  as  senile  plaques.  Aβ  is  believed  to  play  a  central  role  in  AD  by  causing  synaptic  dysfunction  and  cognitive  deficits.  Aβ   derives   from   the   Amyloid   Precursor   Protein   (APP),   which   can   undergo   two   mutually   different  pathways  in  the  cell.  In  the  amyloidogenic  pathway,  APP  is  sequentially  cleaved  by  the  protease  BACE1  and   by   the   γ-­‐secretase   to   produce   Aβ.   In   the   non-­‐amyloidogenic   pathway,   α-­‐secretase   (ADAM10),  cleaves  APP  within  the  Aβ  domain,  thus  preventing  Aβ  generation.  The   correct   spatial   localization  of  ADAM10   in   the  plasma  membrane   is  pivotal   for   an  efficient  APP  α-­‐secretase   cleavage,   thus   the  mechanisms   regulating   the   trafficking   of   ADAM10   play   a   key   role   in   its  activity.  We  have  previously  demonstrated  that  the  interaction  of  ADAM10  cytoplasmic  tail  to  binding  partners,  i.e.   SAP97,   can   modulate   its   localization.   To   identify   new   ADAM10   binding   partners,   which   may   be  involved   in   ADAM10   trafficking/activity   regulation,   we   performed   a   yeast   two-­‐hybrid   screening   of   a  cDNA  brain   library  using  ADAM10  C-­‐terminal   tail  as  a  bait.  Among  the  positive  clones  we   focused  our  attention  on  CAP2.  To   confirm   these   results   we   performed   immunoprecipitation   experiments   and   demonstrated   that  ADAM10   and   CAP2   co-­‐precipitated   from   rat   brain   crude   membrane   fraction.   We   confirmed   this  interaction  by  co-­‐immunoprecipitation  assays  in  a  heterologous  system,  i.e.  HEK293  cells,  where  EGFP-­‐CAP2  and  ADAM10  HA  were  overexpressed.  To   identify   the   domain   responsible   for   the   interaction  we   carried   out   pull-­‐down   assays  with   deletion  mutants  of   the  GST-­‐ADAM10  C-­‐terminal   tail   fusion  protein.  We  demonstrated   that  CAP2  binds   to   the  membrane  proximal  domain  of  ADAM10  cytoplasmic  tail.  In   light   of   the   above,   we   confirmed   that   CAP2   is   a   binding   partner   of   ADAM10.   Further   studies   are  necessary   to   fully   characterize   ADAM10/CAP2   association   and   its   role   in   ADAM10   function   and   AD  pathogenesis.    

     

  23  

 Pathogenetic   role   of   CSF   anti-­‐Aβ   autoantibodies   in   CAA-­‐related   inflammation   and   Alzheimer’s  disease:   a   novel   biomarker   for   the   Amyloid   Related   Imaging   Abnormalities   for   amyloid-­‐modifying  therapies.        Piazza   F.1,   Greenberg   S.M.2,   Savoiardo   M.3,   Giardinetti   M.1,   Chiapparini   L.3,   Raicher   I.4,   Nitrini   R.4,    Sakaguchi  H.5,  Brioschi  M.6,  Billo  G.7,  Colombo  A.8,  Lanzani  F.8,  Piscosquito  G.9,  Carriero  M.R.9,  Giaccone  G.10,  Tagliavini  F.10,  Ferrarese  C.1and  DiFrancesco  J.C.1  

 1  Department  of  Surgery  and  Translational  Medicine,  University  of  Milano-­‐Bicocca,  Monza,  Italy    2  Hemorrhagic  Stroke  Research  Program,  Department  of  Neurology,  Massachusetts  General  Hospital  &  Harvard  Medical  School,  Boston,  MA,  USA  3  Department  of  Neuroradiology,  IRCCS  Foundation  Neurological  Institute  Carlo  Besta,  Milan,  Italy  4  Department  of  Neurology,  University  of  São  Paulo  School  of  Medicine,  São  Paulo,  Brazil  5   Department   of   Neurology,   Graduate   School   of  Medical   Sciences,   Kumamoto   University,   Kumamoto,  Japan  6  Department  of  Neurosciences,  Ospedale  Niguarda  Ca'  Granda,  Milan,  Italy  7  Department  of  Neurology,  St.  Bortolo  Hospital,  Vicenza,  Italy  8  Department  of  Neurology,  Desio  Hospital,  Desio,  Italy  9  Department  of  Cerebrovascular  Diseases,   IRCCS  Foundation  Neurological   Institute  Carlo  Besta,  Milan,  Italy  10   Department   of   Neurodegenerative   Diseases   and   Division   of   Neuropathology-­‐Neurology,   IRCCS  Foundation  Neurological  Institute  Carlo  Besta,  Milan,  Italy        Objective.   Cerebral   Amyloid   Angiopathy-­‐related   inflammation   (CAA-­‐ri)   is   a   rare   meningoencephalitis  characterized   by   vasogenic   edema   and   multiple   cortical/subcortical   microbleeds,   shearing   several  aspects   with   the   recently   defined   Amyloid-­‐Related   Imaging   Abnormalities   (ARIA)   during   Alzheimer's  disease   (AD)   passive   immunization   therapies.   Here,   we   investigated   the   putative   role   of   anti-­‐Aβ  autoantibodies  as  the  primary  actor  of  the  inflammatory  reaction  rising  in  these  two  conditions.      Methods.   Through   a   world   wide   case-­‐control   study   in   80   subjects,   using   a   novel   ultra-­‐sensitive  technique   (patent  application  pending),  we  evaluated  the  anti-­‐Aβ  autoantibodies  concentration   in   the  CSF  of  CAA-­‐ri,  CAA,  AD,  MS  and  healthy-­‐control  subjects.  Levels  of  circulating  Aβ40,  cAβ42,  tau,  P-­‐181  tau  and  APOE4  genotype  were  also  investigated.      Results.   We   confirmed   a   direct   involvement   of   anti-­‐Aβ   autoantibodies   during   the   course   of   CAA-­‐ri,  demonstrating   that   their   concentration   is   specifically   increased   during   the   acute   phase   and  progressively  reduced  with  clinical  and  radiological   remission.  Moreover,  a  strong  correlation  with  the  increased  mobilization  of  cAβ40  and  cAβ42  during  the  acute  phase  was  shown,  followed  by  their  return  to  control  levels  and  reduced  tau  and  P-­‐181  tau  after  remission.      Conclusions.  Our  data  strongly  support  the  hypothesis  that  the  pathogenesis  of  CAA-­‐ri  is  mediated  by  a  selective   autoimmune   reaction   against   cerebro-­‐vascular   Aβ,   directly   related   to   autoantibodies  concentration  and  the  cAβ  overload.  Given  the  similarities  between  spontaneous  ARIA  which  develops  in  CAA-­‐ri  and  that  induced  through  immunization  strategies,  anti-­‐Aβ  autoantibodies  in  the  CSF  may  be  proposed  as   a   valid   alternative   for   the  diagnosis  of  CAA-­‐ri   and  as   a  novel  biomarker   to  monitor  ARIA  during  the  ongoing  amyloid-­‐modifying  therapies  for  AD.      

  24  

 Differential  deregulation  of  astrocytic  calcium  signaling  by  Amyloid  beta,  TNFalpha  and  LPS.      Ronco  Virginia,  Lim  Dmitry  and  Genazzani  Armando  A    Dept.  Pharmaceutical  Sciences,  Amedeo  Avogadro  Univ,  Novara,  Italy.        Alzheimer’s  disease  (AD)  is  a  devastating  neurological  disorder  that  affects  increasing  number  of  people  in   elderly   worldwide.   The   amyloid   beta   (Abeta)   hypothesis   of   AD   is   the  most   popular   and   the  most  studied  one  to  explain  pathogenetic  mechanisms  of  the  disease.  Neuroinflammation  is  an  integral  part  of  the  AD  pathogenesis  and  plays  an  important  role  in  sensitizing  neurons  to  the  toxic  effects  of  amyloid  beta.   Growing   body   of   evidence   suggest   that   reactive   astrocytes,   and   specifically,   deregulation   of  astrocytic   calcium   signaling,   plays   a   pivotal   role   in   the   early   synaptic   dysfunction   and   the   memory  impairment.  Little  (if  anything)  is  known  about  very  early  astrocytic  activation,  whether  it  is  induced  by  toxic  Abeta  peptide  or,  alternatively,  the  pro-­‐inflammatory  environment,  created  by  activated  microglia,  plays  a  pivotal   role.  Previously  we  have  shown  that  Abeta  deregulates  astrocytic  calcium  homeostasis  via   activation   of   calcineurin   (CaN)   and   NF-­‐kB.   We   therefore   treated   primary   cultured   hippocampal  astrocytes  with  nanomolar  [Abeta],  the  major  brain  pro-­‐inflammatory  cytokine  TNFa  and  LPS  as  a  classic  activator   of   glial   cells.   In   stimulated   astrocytes  we   investigated   i)   changes   in  mRNA   levels   of   the   key  components  of  the  astrocytic  calcium  signaling  (mGluR5,  IP3R1  and  IP3R2);   ii)  degradation  of  IkBa  as  a  marker  of  the  NF-­‐kB  activation.  We  report  that  all  three  activating  agents  induced  degradation  of  IkBa  and  therefore  activation  of  NF-­‐kB,  although  there  was  a  clearly  distinct  pattern  of  changes  in  expression  of  the  calcium  signaling  genes.  Specifically:  Abeta  induced  up-­‐regulation  of  all  three  target  genes;  TNFa  down-­‐regulated  mGluR5  and  up-­‐regulated  IP3R2  although  IP3R1  was  unaffected;  LPS  down-­‐regulated  all  three   genes  with   profound  effect   on  mGluR5,   although  down-­‐regulation  of   IP3R1   and   IP3R2  was   less  prominent.    Experiments  are  now  underway  to  investigate  which  of  these  changes  are  mediated  by  CaN-­‐NF-­‐kB  axis  and/or  the  other  signaling  routes  are  involved.      

  25  

 SAP97  is  involved  in  PKC  activation  of  ADAM10  α-­‐secretase.      Saraceno   Claudia1,  Marcello   Elena1,  Musardo   Stefano1,   Pelucchi   Silvia1,   Gardoni   Fabrizio1   and   Di   Luca  Monica1.    1  Department  of  Pharmacological  and  Biomolecular  Sciences,  University  of  Milan,  via  Balzaretti  9,  20133  Milan,  Italy        ADAM10  (a  disintegrin  and  metalloproteinase  10)  is  the  most  accredited  candidate  as  α-­‐secretase  in  the  amyloid  cascade,   since   it  prevents  Aβ   formation.  ADAM10   is   synthesized   in  an   inactive   form,  which   is  proteolytically   activated   during   its   forward   transport   along   the   secretory   pathway   and   at   the   plasma  membrane.  Therefore,  modulation  of  its  trafficking  could  provide  a  mechanism  to  finely  tune  its  activity.  We  reported  that  ADAM10  interacts  directly  with  SAP97,  a  protein  involved  in  trafficking  of  glutamate  receptors,   and   this   interaction   is   required   for   ADAM10   localization   and   enzymatic   activity   at  postsynaptic   membranes.   It   has   been   shown   that   PKC   activation   stimulates   α-­‐secretase   activity   and  inhibits  the  secretion  of  Aβ.  In  particular,  Alzheimer's  disease  patients  have  been  reported  to  have  lower  levels  of  PKC  activity.  Moreover,  PKC  activity  may  regulate  the  subcellular  localization  of  ADAM10.  In  this  framework,  the  aim  of  our  study  is  to  investigate  the  involvement  of  ADAM10/SAP97  complex  in  the  mechanism  underlying  PKC-­‐induced  alpha-­‐secretase  activation.  Here  we  report  that  30  minutes  in  vitro  PKC  activation  via  Phorbol  12,13-­‐Dibutyrate  (PDBu)  induces  an  increase  of  ADAM10  levels  in  the  postsynaptic  compartment  and  a  parallel  reduction  of  enzyme  level  in  the   microsomal   fraction.   To   evaluate   the   role   of   ADAM10/SAP97   complex   in   ADAM10   secretory  trafficking,  we   took   advantage   of   a   cell-­‐permeable   peptide,   Tat-­‐Pro   ADAM10709-­‐729,   which  mimics   the  proline-­‐rich   region   of   ADAM10,   responsible   for   its   association   to   SAP97   and   consequently   interferes  with   ADAM10/SAP97   interaction.   The   treatment   with   the   Tat-­‐Pro   ADAM10709-­‐729   peptide   is   able   to  prevent   PDBu-­‐induced   ADAM10   trafficking   to   the   postsynaptic   compartment,   suggesting   an  involvement   of   SAP97.   A   step   forward   will   be   the   understanding   of   the   mechanism   underlying   this  effect,  which  may  have  a  functional  implication  for  the  regulation  of  alpha-­‐secretase  activity.      

  26  

 Nilotinib   or   Dasatinib,   but   not   Imatinib,   might   reduce   plasma   beta-­‐amyloid   in   leukemia   patients:    implications  for  Alzheimer’s  disease.      Tremolizzo  Lucio,1    Conti  Elisa,1  Zoia  Chiara  Paola,1  Rizzo  Christian,2  Gambacorti-­‐Passerini  Carlo,2  Weksler  Marc  E.,3  Weksler  Babette,3  and  Ferrarese  Carlo1    1Dept.   of   Surgery   and   Interdisciplinary   Medicine,   2Dept.   of   Health   Sciences,   Univ.   of   Milano-­‐Bicocca,  Monza,  Italy;  3Weill  Cornell  Medical  College,  New  York,  USA        Imatinib  Mesylate  is  a  competitive  tyrosine-­‐kinase  inhibitor  (TKI)  used  in  the  treatment  of  multiple  types  of   leukaemia,   including   Ph+   chronic  myelogenous   leukaemia.   The   recent   report   that   Imatinib   inhibits  gamma-­‐secretase   activating   protein   (gSAP),   reducing   beta-­‐amyloid-­‐   (Abeta)   production   without  affecting   Notch-­‐1   cleavage   has   led   to   the   proposal   of   a   possible   application   of   this   drug   in   AD  therapeutics.   Imatinib   peripheral   administration   (20  mg/kg/day)  was   able   to   reduce  Abeta   content   in  mouse  brain.  Usually,  leukaemia  patients  receive  an  average  oral  dose  of  400  mg/day  (~5-­‐6  mg/kg/day).  Other   TKI   used   in   the   field   are   second   generation   compounds   such   as   Nilotinib   and   Dasatinib.   We  recently  reported  in  a  longitudinal  study  that  Imatinib  administration  in  10  leukaemia  patients  was  not  effective   in   reducing   plasma   Abeta1-­‐40   levels.   Here   we   extend   our   observations   including   cross-­‐sectional   data   on   51   leukaemia   patients,   respectively   taking   either,   Imatinib   (n=30),   Dasatinib   or  Nilotinib   (n=9,   D/N),   or   drugs   other   than   TKI   (n=12).   10   healthy   controls   were   included   as   well   and  plasma  Abeta1-­‐40  assessed  by  ELISA  as  previously  described.  D/N  patients  displayed  significantly  lower  values  with  respect  to  all  the  other  groups  (average  reduction  ~30%).  Albeit  our  preliminary  data  do  not  support  a  role  for  Imatinib  in  modulating  Abeta1-­‐40  plasma  levels,  second  generation  TKI  might  be  able  to  do  it.  A  longitudinal  study  on  D/N  patients  is  currently  ongoing  for  testing  this  hypothesis.  

     

  27  

 

ERK  signalling  modulates  APP  metabolism,   tau  phosphorylation  and  EAAT1  processing   in   fibroblasts  from  Alzheimer’s  patients.  

   Zoia   Chiara   Paola*,   Conti   Elisa*,   Costantino   Gisella*,   Susani   Emanuela*,   Isella   Valeria*,   Tremolizzo  Lucio*  and  Ferrarese  Carlo*°    *Laboratory  of  Neurobiology,  Department  of  Surgery  and  Interdisciplinary  Medicine,  School  of  Medicine,  University   of   Milano-­‐Bicocca,   Monza   (MB),   and   °Department   of   Neuroscience   S.   Gerardo   Hospital,  Monza  (MB)        ERK1/2  modulation  was  demonstrated  in  AD  neurons  and  dystrophic  neuritis  (Veeranna,  2004;  Webster  2006)  and  it  is  released  in  parallel  with  tau  and  phospho-­‐tau  in  CSF  patients  (Klafki  2009).  Its  activation  is  involved  in  the  APP  metabolism  and  ERK-­‐1/2-­‐induced  generation  of  Abeta  is  a  significant  signal  for  the  development   of   AD   (Veeranna   2004).   p-­‐ERK1/2   is   associated   with   age-­‐dependent   amyloid   plaque  deposition,  increasing  oxidative  stress,  tau  phosphorylation  and  the  loss  of  synaptophysin  in  presenilin  2  (PS2)   transgenic  mice.   (Lee  2009-­‐2011,  Park  2012).   ERK-­‐pathway   is   also   involved   in   anti-­‐inflammatory  processes  and,  by  specific  transcriptional  factors  (CREB),  might  modulate  the  EAAT1-­‐mRNA.  By  western   blot   and   phospho-­‐Elisa,   fibroblasts   from  AD  patients,  MCI,   and   age-­‐related   subjects  were  tested   to   investigate  ERK-­‐pathway  alterations  associated   to   the  different   stages  of  disease.  APP-­‐alpha  isoforms  were   detected   in   cell   lysates   by   Elisa-­‐kit   (Biosource).   By   EZ-­‐ChIP   assay   (Millipore),   ERK-­‐CREB  association   with   EAAT1-­‐promoter   was   tested   to   explain   the   mRNA   EAAT1   increase,   previously  demonstrated  correlating  with  disease-­‐severity  in  AD  fibroblasts  (r=0.8306;  Zoia  2005).  p70S6  kinase,  a  cytoplasmic   protein   involved   in   both   protein   synthesis   and   phosphorylation   of   Tau,   was   detected   by  western  blot.  Phospho-­‐ERK   was   reduced   by   60%   in   fibroblasts   from   MCI   and   mild-­‐to-­‐moderate   AD,   compared   to  severe  patients  and  controls.  An   inverse  correlation  was  observed  between  phospho-­‐ERK  and  disease-­‐severity   (r=-­‐0.584).   A   50%   reduction   of   APP-­‐alpha   was   shown   in   AD   compared   to   control   subjects  (p=0.03).   MCI   APP-­‐alpha   distribution   was   heterogeneous.   Moreover,   phospho-­‐ERK   and   APP-­‐alpha  correlation  in  control  subject  was  very  significant  (r=0.935),  while  in  MCI  subjects  and  AD  patients  this  correlation  was   lost.   In  AD   fibroblasts,   an   increased  association  between  EAAT1-­‐DNA  and  pCREB,   and  p70S6K  phosphorylation  were  observed,   justifying  a  specific  ERK   involvement   in  EAAT1-­‐mRNA  and  tau  processing.   So,   ERK   pathway  might   help   to   investigate   and  monitor   the   disease   physiopathology   and  progression,  aiming  to  develop  new  pharmacological  strategies  in  AD  fibroblasts,  too.