You are on page 1of 7

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/313426187

Exploration of Various Proteins for the Treatment of Alzheimer's Disease

Article  in  Current Drug Metabolism · February 2017


DOI: 10.2174/1389200218666170203110135

CITATIONS READS
10 127

4 authors, including:

Badar ul Islam Syed Kashif Zaidi


Aligarh Muslim University Center of Excellence In Genomic Medicine Research-KAAU
14 PUBLICATIONS   241 CITATIONS    84 PUBLICATIONS   1,985 CITATIONS   

SEE PROFILE SEE PROFILE

Shams Tabrez
King Abdulaziz University
177 PUBLICATIONS   4,777 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Call for review articles for SI of Medicinal Chemistry View project

Call for submission of your manuscripts to our special issue: "Synopsis on Antimicrobials Challenges—From Dentistry to Environmental Visions" View project

All content following this page was uploaded by Shams Tabrez on 10 April 2018.

The user has requested enhancement of the downloaded file.


808 Send Orders for Reprints to reprints@benthamscience.ae

Current Drug Metabolism, 2017, 18, 808-813


REVIEWARTICLE
ISSN: 1389-2002
eISSN: 1875-5453

Current
Drug
Metabolism
Impact
Factor:
2.659

Exploration of Various Proteins for the Treatment of Alzheimer’s Disease The international

journal for timely

in-depth reviews

on Drug Metabolism

BENTHAM
SCIENCE

Badar ul Islam1, Syed Kashif Zaidi2, Mohammad Amjad Kamal3,4 and Shams Tabrez3,*

1
Department of Biochemistry, J. N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, India; 2 Cen-
ter of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; 3King Fahd Medical Re-
search Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; 4 Enzymoics, 7 Peterlee Place, Hebersham, NSW, Australia

Abstract: Background: Alzheimer’s disease (AD) is an irreversible multifaceted neurodegenerative disorder that
gradually degrades neuronal cells. It is the most frequent cause of memory loss and dementia in elderly individuals
worldwide. The extracellular deposition of beta amyloid (A), intracellular neurofibrillary tangles (NFTs) retention,
ARTICLE HISTORY neuronal decline and neurotransmitter system derangement are the patho-physiological marker of this devastated
Received: September 12, 2016 disease.

tio y
Revised: October 06, 2016 Objective: In view of limited treatment option and their success rate, there is an urgent need to explore the vast array
Accepted: November 11, 2016
of proteomes for the management of AD. These proteins could be therapeutically targeted to prevent the progression

bu nl
DOI: of this disease. In the present review, we tried to uncover several proteins that could be exploited in AD therapeutics.

n
10.2174/1389200218666170203110135
Conclusion: Based on our article, we conclude that proteome based AD treatment needs more refinements and

tri O
approaches to achieve the desired success rate.
Keywords: Alzheimer’s disease, amyloid beta, amyloid precursor protein, apolipoprotein, tau, therapeutics.
is e
1. INTRODUCTION different proteins that could be therapeutically targeted to decipher
s
A multifactorial neurodegenerative disease (Alzheimer’s) the mysteries of AD puzzle in clinical perspectives.
rD U

gained interest among researchers for the past several years due to
1.1. Amyloid Precursor Protein (APP)
its worldwide etiology [1]. It is the most common form of dementia
Current Drug Metabolism

characterized by neuronal dysfunction and apoptosis that results in Amyloid precursor protein is a type I transmembrane protein. It is
Fo al

memory decline and impaired cognitive functions. About 36 million produced in the endoplasmic reticulum (ER) as an antecedent for A
people across the globe are affected by this disorder and this num- and other peptides [15]. Post-translation, it is extensively altered via
ber is expected to reach up to 114 million by 2050 [2, 3]. AD is glycosylation followed by phosphorylation at some residues inside
ot on

categorized into early onset or familial (1-6%) and sporadic (>90%) the cytoplasmic domain [16]. In this domain the interaction with vari-
[4]. Its multifaceted pathophysiology includes enhanced retention ous cytoplasmic proteins took place which aids in transport to differ-
of amyloid beta (A) plaque around neuronal cells and neurofibril- ent locations [17]. In amyloidogenic pathway, APP is cleaved by -
N rs

lary tangles formed by hyper-phosphorylated tau-related microtu- and -secretase enzymes whereas in non-amyloidogenic pathway,
bule inside cells [5]. Some other disturbed mechanisms like calcium cleavage is carried out by - and -secretase enzymes (Fig. 1). The
Pe

release, persistent oxidative imbalance, mitochondrial and mitotic soluble APP fragment has been noted for neuroprotective features
dysfunction, hormonal disparity and inflammation are also believed and signaling properties [18, 19]. Immunocytochemical studies re-
to participate in AD pathogenesis. The proteasome system is the vealed the locations of APP from the ER to lysosomes and auto-
main clearance machinery for hyper-phosphorylated tau protein phagosomes [20]. Both full-length and fragmented APP exist at sev-
which gets blocked by A oligomers and plaques [6]. As a result, eral unusual locations viz. nucleus [21], the ciliary root of retinal
intracellular Ca2+ concentration gets disturbed which activates vari- photoreceptor cells [22], mitochondria [23], and cytoplasm [24]. APP
ous inflammatory cascades, thereby promote neuronal dysfunction intracellular domain (APP fragment) can either enter the nucleus
[7, 8]. Different hypotheses for AD pathogenesis have been put alone or in combination with Fe65 and APP binding protein [21].
forwarded in literature which includes amyloid [9], glutamate- This localization mode suggests a novel target that could aid in AD
mediated or glutamatergic [10], oxidative imbalance [11], inflam- therapy. Other widely used strategies for AD therapy are the preven-
matory-mediated [12], acetylcholine-mediated or cholinergic [13] tion of A generation, improvement in proper APP processing
and metal-based [14]. through inhibition of -, - or -secretase enzymes [25, 26].
The complex mechanisms involved in AD progression restrain 1.2. Amyloid Beta (A)
the actual beneficial effect associated with any anti-Alzheimer drug.
Till date, memantine, and four other cholinesterase inhibitors are Amyloid beta peptides are formed by the action of various se-
approved by the US-FDA (Food and Drug Administration) for AD cretase enzymes which lead to the formation of amyloid plaques
management. However, these drugs were unable to divert the route [27]. Numerous studies demonstrated lower A42 density in cere-
of the disease rather they just provide symptomatic effects. There- bro spinal fluid (CSF) of AD patients [28, 29]. This notion is not
fore, the need of the hour is to find out novel and sophisticated clearly understood but it is believed that A42 confiscation to
therapeutic alternatives. In the present review, we focus on the plaques is the main reason behind the reduced clearance of A into
CSF [30]. On the other hand, Creuztfeldt-Jakob disease patients
also have decreased levels of A clearance but they did not have
*Address correspondence to this author at the King Fahd Medical Research any amyloid plaque deposition [31]. Hence, the decreased clearance
Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi of A is believed to be due to the different mechanisms that take
Arabia; E-mail: shamstabrez1@gmail.com place in various diseases. A recent study provided a notion that

1875-5453/17 $58.00+.00 © 2017 Bentham Science Publishers


Exploration of Various Proteins for the Treatment of Alzheimer’s Disease Current Drug Metabolism, 2017, Vol. 18, No. 9 809

NON-AMYLOIDOGENIC PATHWAY

a-Secretase g-Secretase P3 Peptides

Amyloid Precursor Protein (APP)

b-Secretase g-Secretase Amyloid Beta (Ab) Ab Oligomers Ab Fibrils Ab Plaques


monomers
AMYLOIDOGENIC PATHWAY

Fig. (1). Amyloidogenic and non-amyloidogenic pathway depicting APP cleavage by secretase enzymes.

decreased A42 clearance in AD patients is most probably due to employing somatostatin or its analogs to target neprilysin
the accumulation of A42 monomers into oligomers that results could be a good therapeutic approach in AD therapy. RAGE-
into epitope camouflaging accompanied by conformational altera- mediated active transport is the target of interest for the scien-
tions and making them inaccessible to antibodies [32]. Furthermore, tists as it transports A across the blood-brain barrier and aid
this assumption is supported by the oligomerization of A42 in the in their deposition. Hence the use of RAGE inhibitors is con-
brain parenchyma of AD patients. sidered as another therapeutic option in AD therapy. One of
The possible A targeting approaches are listed below: RAGE inhibitors (PF-04494700) has successfully completed
phase 2 trials. Different monoclonal antibodies have the ability
Reducing A Production: It can be achieved either by the activating to bind A and aid in their clearance via microglial phagocyto-
-secretase or inhibiting - and -secretase enzymes.

tio y
sis [59]. Solanezumab (LY2062430), a humanized monoclonal
Arresting A Oligomerization or Fibrillization: A peptides have antibody against A16-24, binds with soluble A [60] whereas

bu nl
the ability to self-aggregate that leads to the formation of oli- gantenerumab (R1450, a monoclonal antibody against A1-

n
gomers, protofibrils, fibrils and finally A plaques [33, 34]. A 11) and crenezumab (humanized monoclonal antibody against

tri O
vast array of molecules has been recognized that could either A40/A42) have been reported to decrease cerebral A
arrest A aggregation and fibrillogenesis or hinder A toxicity. through microglial phagocytosis [61, 62].
Congo red (A fibril binding dye) has been reported to inhibit
is e
fibril generation along with their beneficial effect on neurotox- 1.3. Tau Protein
icity [35, 36]. Other compounds such as sulfonated anions, sul- Tau is a microtubule-associated protein, commonly expressed
s
fonated dyes (benzofuran-type compounds) and amphiphilic in neurons, where it primarily takes part in microtubule stabilization
rD U

surfactants (di-C6-PC and di-C7-PC) have also been reported (comprising nucleation, growth, and bundling), thereby regulates
for A inhibiting potential [37-39]. One study reported clear- functional organization of the neuronal cells [63]. C-terminal end of
ance of amyloid plaque by active immunization with A (AN tau protein contains microtubule-binding domain that is involved in
Fo al

1792) in AD patients [40]. Tramiprosate (glycosaminoglycan) the polymerization and stabilization of microtubules [64]. Whereas
arrest the A oligomerization and fibril formation by interact- N-terminal is composed of highly acidic amino acids followed by
ing with A monomers while bapineuzumab (humanized
ot on

proline-rich area. This domain is involved in the interaction be-


monoclonal antibody against A1-5) interact with both soluble tween cytoskeleton and plasma membrane [65]. Phosphorylated tau
and fibrillar forms of A that decreases amyloid load in trans- interacts with microtubules with reduced affinity resulting in the
genic mice [41-43]. Another compound ELND005 or scyllo- instability of microtubules. This reduction in microtubule stability
N rs

inositol also inhibits A oligomerization by reducing soluble leads them into filamentous and non-filamentous aggregates that
A, hence reverses the cognitive decline in transgenic mice convert into neurotoxic NFTs and neuritic plaques at later stage
Pe

[44]. Colostrinin (a proline-rich peptide) has been reported for [66]. The focal point of AD pathogenesis is tau hyperphosphoryla-
its anti-aggregation property that results in the dissolution of tion that causes microtubule instability [67].
pre-formed fibrils [45]. Recently, several new peptides with
Tau targeting strategies are depicted in Fig. (2) and are also
anti-aggregation potential have been synthesized [46]. An ac-
listed below:
tin-binding protein, gelsolin plays an essential role in the as-
sembly and disassembly of actin filaments and is extracellu- Preventing Tau Phosphorylation: Kinase blocking is one of the
larly found in CSF and plasma and intracellularly in mito- convincing strategy to prevent tau phosphorylation. Various
chondria and cytosol [47]. It has the ability to hamper A fi- tau kinases capable of phosphorylating tau are cyclin-
brillization and helps in the dissolution of preformed A fi- dependent protein kinase 5 (cdk5), glycogen synthase kinase
brils, which increases its clearance [48]. 3 (GSK-3), stress-activated protein kinase (SAPK), micro-
Increasing A Clearance: Several proteases, such as angiotensin- tubule affinity-regulating kinase (MARK), eukaryotic transla-
converting enzyme (ACE), insulin degrading enzyme (IDE), tion initiation factor 2 alpha kinase 2 (EIF2AK2), dual-
cathepsin B, metalloproteinase 9, neprilysin and plasmin, play specificity tyrosine (Y)-phosphorylation regulated kinase 1A
a major role in the digestion of A plaques and their decline (DYRK1A), A-kinase anchor protein 13 (AKAP-13) and
may lead to retention of A in AD patients [49-51]. ACE has cAMP-dependent protein kinase (PKA) have been listed in lit-
been reported to notably arrest A aggregation, retention, and erature [68-70]. Most studies reported lithium as the main
cytotoxicity [52]. IDE is expressed in almost all tissues but blocker of GSK-3 however, several other compounds such as
greatly in testis, brain, liver and muscle and has A digesting sodium valproate, flavopiridol, aloisines, aminothiazole AR-
capability [53-55]. Therefore, strategies that could increase the A014418, pyrazolopyridines and pyrazolopyrazines have also
expression and activity of IDE could also be exploited as a po- been reported for GSK-3 blocking activity. Likewise, cdk5
tential approach for AD therapy. Cathepsin B (a cysteine pro- inhibitors like indirubins, purine olomoncine, aloisines and
tease), appreciably decreases A deposition through degrada- flavopiridol are also considered for this purpose [69, 71].
tion of A1-42 at C-terminal and believed to reduce its level in Moreover, several important signaling pathways are actively
AD brain [56, 57]. Somatostatin, a peptide hormone, modu- regulated by the kinases, therefore one should consider the
lates the A removal via neprilysin activation [58]. Therefore, possible side effects caused by the use of kinase blockers [72,
810 Current Drug Metabolism, 2017, Vol. 18, No. 9 Islam et al.

Tau Targeting Strategies

Prevention of tau Prevention of microtubule Block tau oligomerization or Increasing tau digestion Increasing tau
phosphorylation destabilization by the use of aggregation by the use of clearance
1. Blockage of various 1. Paclitaxel 1. Hsp90 inhibitors 1. Immunological-based
kinases (cdk5, GSK-3b, 1. Tau-binding drugs such as such as curcumin approach such as use
SAPK, MARK, etc.) lansoprazole and astemizole of monoclonal
2. Epothilone D
2. Activation of phosphatases antibodies against
by drugs such as metformin 3. Neuropeptides such as 2. Methylene blue phosphorylated tau
and sodium selenite NAP and D-SAL
3. Natural phenolic
compounds
(hydroxytyrosol, oleuropein,
oleurpein aglycone, etc.)

Fig. (2). A representative diagram highlighting strategies for tau-targeting.

Table 1. Management of AD through various protein targeting and their molecular participants.

Targeted protein Strategies Participants References

tio y
Amyloid precursor protein (APP) Prevention in amyloid beta (A) generation. Inhibition of - or -secretase enzymes. [25, 26]

bu nl
Improvement in proper APP processing. Inhibition of -, - or -secretase enzymes. [25, 26]

n
tri O
Amyloid beta (A) Reduction in A production. Activation of -secretase or inhibition of [30, 32]
- and -secretase enzymes.

Arresting A oligomerization. Tramiprosate, ELND005 and colostrinin. [41, 44-45]


is e
Arresting A fibrillization. Congo red, gelsolin, bapineuzumab and [35, 37-39,
s
amphiphilic surfactants. 42, 43, 48]
rD U

Increase in A clearance via digestion of A. Angiotensin-converting enzyme, insulin [49-51]


degrading enzyme, cathepsin B,
Fo al

metalloproteinase 9, neprilysin and plasmin.


ot on

Increase in A clearance via neprilysin activation. Somatostatin. [58]

Increase in A clearance via active transport. RAGE inhibitors. [59]


N rs

Increase in A clearance via microglial phagocytosis. Solanezumab, gantenerumab and crenezumab. [61, 62]

Tau protein Prevention in tau phosphorylation by Lithium, sodium valproate, flavopiridol, [69, 71]
Pe

kinase inhibitors. indirubins, purine olomoncine, aloisines etc.

Prevention in tau phosphorylation by phosphatase Metformin, sodium selenite and memantine. [74, 75]
activators.

Reduction in microtubule destabilization. Paclitaxel, epothilone D, and neuropeptides. [76-78]

Prevention in tau oligomerization/aggregation. Lansoprazole, astemizole, methylene blue [79-84]


and natural phenolic compounds.

Acceleration in tau digestion. Hsp90 inhibitors. [86, 87]

Increase in tau clearance. Monoclonal antibodies against [89, 90]


phosphorylated tau.

73]. Another strategy to dephosphorylate tau is the activation tials, such as improvement in microtubule density, motor func-
of protein phosphatases. The key enzyme involved in tau- tion, and axonal transport. Another compound, epothilone D
based dephosphorylation is protein phosphatase 2A (PP2A), has also been noted for microtubule stabilization effects be-
thus its activators are believed to have therapeutical potential sides blood-brain barrier clearance potential [76]. Neuropep-
in AD treatment. Drugs, such as metformin and memantine tides, such as NAP and D-SAL, have also been documented
have been reported for PP2A activating properties [74, 75]. On for boosting effect on microtubule stabilization [77, 78].
the other hand, sodium selenite (Ve-015), a PP2A activator, is Blocking Tau Oligomerization/Aggregation: One of the important
under clinical investigation. tau-based AD treatment approaches is to find out the com-
Reduction in Microtubule Destabilization: Paclitaxel (a strong anti- pounds that could block tau interaction and NFT deposition.
cancer drug), is reported for microtubule stabilization poten- Tau-binding drugs, such as lansoprazole and astemizole, are
Exploration of Various Proteins for the Treatment of Alzheimer’s Disease Current Drug Metabolism, 2017, Vol. 18, No. 9 811

indirectly helpful in reducing tau-tau interactions thereby ar- [7] Lacor, P.N.; Buniel, M.C.; Furlow, P.W.; Clemente, A.S.; Velasco,
resting tau oligomerization/aggregation [79]. Methylene blue P.T.; Wood, M.; Viola, K.L.; Klein, W.L. Abeta oligomer-induced
has been documented for various effects that include preven- aberrations in synapse composition, shape, and density provide a
tion of tau interactions, amyloid aggregation, acetylcholine es- molecular basis for loss of connectivity in Alzheimer’s disease. J.
Neurosci., 2007, 27, 796-807.
terase inhibition, reduction in oxidative stress, improvement in [8] Wenk, G.L. Neuropathologic changes in Alzheimer’s disease. J.
electron transport and mitochondrial damage aversion [80-83]. Clin. Psychiat., 2003, 64, S7-S10.
Daccache et al. [84] reported naturally occurring phenolic [9] Goedert, M.; Spillantini, M.G. A century of Alzheimer's disease.
compounds (hydroxytyrosol, oleuropein and its derivative Science, 2006, 314, 777-781.
oleuropein aglycone) with tau arresting properties [84]. [10] Bezprozvanny, I.; Mattson, M.P. Neuronal calcium mishandling
Accelerating Tau Digestion: Tau breakage is also considered as a and the pathogenesis of Alzheimer's disease. Trends Neurosci.,
2008, 31, 454-463.
promising treatment option against AD pathogenesis. Chaper- [11] Praticò, D. Evidence of oxidative stress in Alzheimer's disease
ones, like Hsp90, involved in the prevention of tau breakage brain and antioxidant therapy: Lights and shadows. Ann. N. Y.
besides its role in proper folding of denatured proteins [85]. Acad. Sci., 2008, 1147, 70-78.
Curcumin (Hsp90 inhibitors) has also been reported for tau [12] Trepanier, C.H.; Milgram, N.W. Neuroinflammation in Alzheimer's
breaking potential [86, 87]. However, the only concern related disease: are NSAIDs and selective COX-2 inhibitors the next line
with chaperone blocking is the probable intrusion in their main of therapy? J. Alzheimers Dis., 2010, 21, 1089-1099.
activity may cause adverse effects. [13] Craig, L.A.; Hong, N.S.; McDonald, R.J. Revisiting the cholinergic
hypothesis in the development of Alzheimer's disease. Neurosci.
Increasing Tau Clearance: Immunological based approach for Biobehav. Rev., 2011, 35, 1397-1409.
clearance of tau and its related fibrils receives interest lately [14] Bonda, D.J.; Lee, H.G.; Blair, J.A.; Zhu, X.; Perry, G.; Smith, M.A.
[88]. Scientific studies reported beneficial effects in tau trans- Role of metal dyshomeostasis in Alzheimer's disease. Metallomics,
genic mice when they were passively immunized with mono- 2011, 3, 267-270.

tio y
clonal antibodies against phosphorylated tau [89, 90]. This [15] Selkoe, D.J. The cell biology of beta-amyloid precursor protein and
suggests a potential of immune based approach for the AD presenilin in Alzheimer's disease. Trends Cell Biol., 1998, 8, 447-

bu nl
treatment. We have also summarized various protein targeting 453.

n
approaches in the Table (1). [16] Walter, J.; Haass, C. Posttranslational modifications of amyloid

tri O
precursor protein: Ecto domain phosphorylation and sulfation.
CONCLUSION Methods Mol. Med., 2000, 32, 149-168.
[17] Muresan, V.; Muresan, Z. Is abnormal axonal transport a cause, a
In view of the current article and above-mentioned literature, contributing factor or a consequence of the neuronal pathology in
is e
we emphasize the exploitation of various proteins and their novel Alzheimer's disease? Future Neurol., 2009, 4, 761-773.
inhibitors for the clinical treatment of AD. The pathogenesis of AD [18] Kojro, E.; Gimpl, G.; Lammich, S.; Marz, W.; Fahrenholz, F. Low
s
is highly complex and several factors and signaling mechanisms cholesterol stimulates the nonamyloidogenic pathway by its effect
rD U

actively participate in it. FDA-approved treatments for AD fail to on the alpha-secretase ADAM 10. Proc. Natl. Acad. Sci. USA,
stop or slow-down disease progression and are related to adverse 2001, 98, 5815-5820.
[19] Selkoe, D.J.; Schenk, D. Alzheimer's disease: molecular under-
side effects that decrease patient interest to therapy. Hence, its standing predicts amyloid-based therapeutics. Annu. Rev. Pharma-
Fo al

treatment still needs new refinements and approaches for novel col. Toxicol., 2003, 43, 545-584.
targets in search to achieve complete endeavor. [20] Xiao, Q.; Yan, P.; Ma, X.; Liu, H.; Perez, R.; Zhu, A.; Gonzales,
ot on

E.; Tripoli, D. L.; Czerniewski, L.; Ballabio, A.; Cirrito, J. R.; Di-
CONSENT FOR PUBLICATION wan, A.; Lee, J. M. Neuronal-targeted TFEB accelerates lysosomal
Not applicable. degradation of APP, reducing A generation and amyloid plaque
pathogenesis. J. Neurosci., 2015, 35, 12137-12151.
N rs

CONFLICT OF INTEREST [21] Cao, X.; Südhof, T.C. A transcriptionally [correction of transcrip-
tively] active complex of APP with Fe65 and histone acetyltrans-
The authors declare no conflict of interest, financial or other-
Pe

ferase Tip60. Science, 2001, 293, 115-120.


wise. [22] Yang, J.; Li, T. The ciliary rootlet interacts with kinesin light
chains and may provide a scaffold for kinesin-1vesicular cargos.
ACKNOWLEDGEMENTS Exp. Cell Res., 2005, 309, 379-389.
The authors gratefully acknowledge the research facility pro- [23] Anandatheerthavarada, H.K.; Biswas, G.; Robin, M.A.; Avadhani,
vided by the Aligarh Muslim University, Aligarh, India and King N. G. Mitochondrial targeting and a novel transmembrane arrest of
Fahd Medical Research Center (KFMRC), King Abdulaziz Univer- Alzheimer's amyloid precursor protein impairs mitochondrial func-
tion in neuronal cells. J. Cell Biol., 2003, 161, 41-54.
sity, Jeddah, Saudi Arabia. Thanks are also due to Mohammad S [24] Takahashi, R.H.; Milner, T.A.; Li, F.; Nam, E.E.; Edgar, M.A.;
Gazdar (Librarian, KFMRC) for providing assistance with the Yamaguchi, H.; Beal, M.F.; Xu, H.; Greengard, P.; Gouras, G.K.
literature. Intraneuronal Alzheimer a beta 42 accumulates in multivesicular
bodies and is associated with synaptic pathology. Am. J. Pathol.,
REFERENCES 2002, 161, 1869-1879.
[1] Ballard, C.; Gauthier, S.; Corbett, A.; Brayne, C.; Aarsland, D.; [25] Schenk, D.; Basi, G.S.; Pangalos, M.N. Treatment strategies target-
Jones, E. Alzheimer's disease. Lancet, 2011, 377, 1019-1031. ing amyloid -protein. Cold Spring Harb. Perspect. Med., 2012, 2,
[2] Mullane, K.; Williams, M. Alzheimer's therapeutics: Continued a00638.
clinical failures question the validity of the amyloid hypothesis-but [26] Saido, T.; Leissring, M.A. Proteolytic degradation of amyloid -
what lies beyond? Biochem. Pharmacol., 2013, 85, 289-305. protein. Cold Spring Harb. Perspect. Med., 2012, 2, a006379.
[3] Silva, T.; Reis, J.; Teixeira, J.; Borges, F. Alzheimer's disease, [27] Kang, J.; Lemaire, H.G.; Unterbeck.; Salbaum, J.M.; Masters, C.L.;
enzyme targets and drug discovery struggles: from natural products Grzeschik, K.H.; Multhaup, G.; Beyreuther, K.; Müller-Hill, B.
to drug prototypes. Ageing Res. Rev., 2014, 15, 116-145. The precursor of Alzheimer's disease amyloid A4 protein resembles
[4] Alzheimer’s Association. 2015 Alzheimer's disease facts and fig- a cell-surface receptor. Nature, 1987, 325, 733-736.
ures. Alzheimers Dement., 2015, 11, 332-384. [28] Blennow, K.; De Leon, M.J.; Zetterberg, H. Alzheimer’s disease.
[5] Noble, W.; Hanger, D.P.; Miller, C.C.J.; Lovestone, S. The impor- Lancet, 2006, 368, 387-403.
tance of tau phosphorylation for neurodegenerative diseases. Front. [29] Shaw, L.M.; Vanderstichele, H.; Knapik-Czajka, M.; Figurski, M.;
Neurol., 2013, 4, 83. Coart, E.; Blennow, K.; Soares, H.; Simon, A.J.; Lewczuk, P.;
[6] De-Paula, V.J.; Radanovic, M.; Diniz, B.S.; Forlenza, O.V. Alz- Dean, R.A.; Siemers, E.; Potter, W.; Lee, V.M.; Trojanowski, J.Q.
heimer’s disease. Subcell. Biochem., 2012, 65, 329-352. Alzheimer's disease neuroimaging initiative. qualification of the
812 Current Drug Metabolism, 2017, Vol. 18, No. 9 Islam et al.

analytical and clinical performance of CSF biomarker analyses in [49] Nalivaeva, N.N.; Beckett, C.; Belyaev, N.D.; Turner, A.J. Are
ADNI. Acta. Neuropathol., 2011, 121, 597-609. amyloid-degrading enzymes viable therapeutic targets in Alz-
[30] Fagan, A.M.; Mintun, M.A.; Mach, R.H.; Lee, S.Y.; Dence, C.S.; heimer’s disease? J. Neurochem., 2012, 120 (Suppl. 1), 167-185.
Shah, A.R.; LaRossa, G.N.; Spinner, M.L.; Klunk, W.E.; Mathis, [50] Barage, S.H.; Sonawane, K.D. Amyloid cascade hypothesis:
C.A.; DeKosky, S.T.; Morris, J.C.; Holtzman, D.M. Inverse rela- Pathogenesis and therapeutic strategies in Alzheimer's disease.
tion between in vivo amyloid imaging load and cerebrospinal fluid Neuropeptides, 2015, 52, 1-18.
Abeta42 in humans. Ann. Neurol., 2006, 59, 512-519. [51] Yasojima, K.; McGeer, E.G.; McGeer, P.L. Relationship between
[31] Wiltfang, J.; Esselmann, H.; Smirnov, A.; Bibl, M.; Cepek, L.; beta amyloid peptide generating molecules and neprilysin in Alz-
Steinacker, P.; Mollenhauer, B.; Buerger, K.; Hampel, H.; Paul, S.; heimer disease and normal brain. Brain Res., 2001, 919, 115-121.
Neumann, M.; Maler, M.; Zerr, I.; Kornhuber, J.; Kretzschmar, H. [52] Hu, J.; Igarashi, A.; Kamata, M.; Nakagawa, H. Angiotensin-
A.; Poser, S.; Otto, M. Beta-amyloid peptides in cerebrospinal fluid converting enzyme degrades Alzheimer amyloid beta-peptide (A
of patients with Creutzfeldt-Jakob disease. Ann. Neurol., 2003, 54, beta); retards A beta aggregation, deposition, fibril formation; and
263-267. inhibits cytotoxicity. J. Biol. Chem., 2001, 276, 47863-47868.
[32] Wang-Dietrich, L.; Funke, S. A.; Kuhbach, K.; Wang, K.; [53] Kuo, W.L.; Montag, A.G.; Rosner, M.R. Insulin-degrading enzyme
Besmehn, A.; Willbold, S.; Cinar, Y.; Bannach, O.; Birkmann, E.; is differentially expressed and developmentally regulated in various
Willbold, D. The amyloid-beta oligomer count in cerebrospinal rat tissues. Endocrinology, 1993, 132, 604-611.
fluid is a biomarker for Alzheimer’s disease. J. Alzheimers Dis., [54] Vekrellis, K.; Ye, Z.; Qiu, W. Q.; Walsh, D.; Hartley, D.;
2013, 34, 985-994. Chesneau, V. Neurons regulate extracellular levels of amyloid beta-
[33] Xu, Y.; Shen, J.; Luo, X.; Zhu, W.; Chen, K.; Ma, J.; Jiang, H. protein via proteolysis by insulin-degrading enzyme. J. Neurosci.,
Conformational transition of amyloid beta-peptide. Proc. Natl. 2000, 20, 1657-1665.
Acad. Sci. USA, 2005, 102, 5403-5407. [55] Farris, W.; Mansourian, S.; Chang, Y.; Lindsley, L.; Eckman, E.
[34] Ahmed, M.; Davis, J.; Aucoin, D.; Sato, T.; Ahuja, S.; Aimoto, S.; A.; Frosch, M.P.; Eckman, C.B.; Tanzi, R.E.; Selkoe, D.J.;
Elliott, J.I.; Van Nostrand, W. E.; Smith, S.O. Structural conversion Guenette, S. Insulin-degrading enzyme regulates the levels of insu-
of neurotoxic amyloid-1-42 oligomers to fibrils. Nat. Struct. Mol. lin, amyloid beta-protein, and the beta-amyloid precursor protein

tio y
Biol., 2010, 17, 561-567. intracellular domain in vivo. Proc. Natl. Acad. Sci. USA, 2003, 100,
[35] Lorenzo, A.; Yankner, B.A. Beta-amyloid neurotoxicity requires 4162-4167.

bu nl
fibril formation and is inhibited by Congo red. Proc. Natl. Acad. [56] Mueller-Steiner, S.; Zhou, Y.; Arai, H.; Roberson, E.D.; Sun, B.;

n
Sci. USA, 1994, 91, 12243-12247. Chen, J.; Wang, X.; Yu, G.; Esposito, L.; Mucke, L.; Gan, L.

tri O
[36] De Felice, F.G.; Ferreira, S.T. Beta-amyloid production, aggrega- Antiamyloidogenic and neuroprotective functions of cathepsin B:
tion, and clearance as targets for therapy in Alzheimer's disease. implications for Alzheimer's disease. Neuron, 2006, 51, 703-714.
Cell Mol. Neurobiol., 2002, 22, 545-563. [57] Sun, B.; Zhou, Y.; Halabisky, B.; Lo, I.; Cho, S. H.; Mueller-
[37] Kisilevsky, R.; Lemieux, L.J.; Fraser, P.E.; Kong, X.; Hultin, P.G.; Steiner, S.; Devidze, N.; Wang, X.; Grubb, A.; Gan, L. Cystatin C-
is e
Szarek, W.A. Arresting amyloidosis in vivo using small-molecule cathepsin B axis regulates amyloid beta levels and associated neu-
anionic sulphonates or sulphates: implications for Alzheimer's dis- ronal deficits in an animal model of Alzheimer's disease. Neuron,
s
ease. Nat. Med., 1995, 1, 143-148. 2008, 60, 247-257.
rD U

[38] Allsop, D.; Howlett, D.; Christie, G.; Karran, E. Fibrillogenesis of [58] Saito, T.; Iwata, N.; Tsubuki, S.; Takaki, Y.; Takano, J.; Huang, S.
beta-amyloid. Biochem. Soc. Trans., 1998, 26, 459-463. M.; Suemoto, T.; Higuchi, M.; Saido, T. C. Somatostatin regulates
[39] Wang, S.S.; Chen, Y.T.; Chou, S.W. Inhibition of amyloid fibril brain amyloid beta peptide Abeta42 through modulation of prote-
formation of beta-amyloid peptides via the amphiphilic surfactants. olytic degradation. Nat. Med., 2005, 11, 434-439.
Fo al

Biochim. Biophys. Acta., 2005, 1741, 307-313. [59] Wisniewski, T.; Konietzko, U. Amyloid-beta immunisation for
[40] Serrano-Pozo, A.; William, C. M.; Ferrer, I.; Uro-Coste, E.; Alzheimer's disease. Lancet Neurol., 2008, 7, 805-811.
ot on

Delisle, M.B.; Maurage, C.A.; Hock, C.; Nitsch, R.M.; Masliah, E.; [60] Teich, A. F.; Arancio, O. Is the amyloid hypothesis of Alzheimer’s
Growdon, J.H.; Frosch, M.P.; Hyman, B.T. Beneficial effect of disease therapeutically relevant? Biochem. J., 2012, 446, 165-177.
human anti-amyloid-beta active immunization on neurite morphol- [61] Ostrowitzki, S., Deptula, D.; Thurfjell, L.; Barkhof, F.; Bohrmann,
ogy and tau pathology. Brain, 2010, 133, 1312-1327. B.; Brooks, D.J.; Klunk, W.E.; Ashford, E.; Yoo, K.; Xu, Z.X.;
N rs

[41] Wright, T.M. Tramiprosate. Drugs Today (Barc), 2006, 42, 291- Loetscher, H.; Santarelli, L. Mechanism of amyloid removal in pa-
298. tients with Alzheimer disease treated with gantenerumab. Arch.
Pe

[42] Teich, A.F.; Arancio, O. Is the amyloid hypothesis of Alzheimer’s Neurol., 2012, 69, 198-207.
disease therapeutically relevant? Biochem. J., 2012, 446, 165-177. [62] Adolfsson, O.; Pihlgren, M.; Toni, N.; Varisco, Y.; Buccarello, A.
[43] Bard, F.; Cannon, C.; Barbour, R.; Burke, R.L.; Games, D,; Gra- L.; Antoniello, K.; Lohmann, S.; Piorkowska, K.; Gafner, V.; At-
jeda, H.; Guido, T.; Hu, K.; Huang, J.; Johnson-Wood, K.; Khan, wal, J. K.; Maloney, J.; Chen, M.; Gogineni, A.; Weimer, R.M.;
K.; Kholodenko, D.; Lee, M.; Lieberburg, I.; Motter, R.; Nguyen, Mortensen, D.L.; Friesenhahn, M.; Ho, C.; Paul, R.; Pfeifer, A.;
M.; Soriano, F.; Vasquez, N.; Weiss, K.; Welch, B.; Seubert, P.; Muhs, A.; Watts, R.J. An effector-reduced anti--amyloid (A) an-
Schenk, D.; Yednock, T. Peripherally administered antibodies tibody with unique A binding properties promotes neuroprotection
against amyloid beta-peptide enter the central nervous system and and glial engulfment of A. J. Neurosci., 2012, 32, 9677-9689.
reduce pathology in a mouse model of Alzheimer disease. Nat. [63] Buée, L.; Bussière, T.; Buée-Scherrer, V.; Delacourte, A.; Hof, P.
Med., 2000, 6, 916-919. R. Tau protein isoforms, phosphorylation and role in neurodegen-
[44] DaSilva, K.A.; Brown, M.E.; Cousins, J.E.; Rappaport, R.V.; Au- erative disorders. Brain Res. Brain Res. Rev., 2000, 33, 95-130.
bert, I.; Westaway, D.; McLaurin, J. scyllo-inositol (ELND005) [64] Goedert, M.; Spillantini, M.G.; Potier, M.C.; Ulrich, J.; Crowther,
ameliorates amyloid pathology in an aggressive mouse model of R. A. Cloning and sequencing of the cDNA encoding an isoform of
Alzheimer’s disease. Alzheimer’s Demen., 2009, 5, P425. microtubule-associated protein tau containing four tandem repeats:
[45] Janusz, M.; Woszczyna, M.; Lisowski, M.; Kubis, A.; Macala, J.; Differential expression of tau protein mRNAs in human brain.
Gotszalk, T.; Lisowski, J. Ovine colostrum nanopeptide affects EMBO J., 1989, 8, 393-399.
amyloid beta aggregation. FEBS Lett., 2009, 583, 190-196. [65] Brandt, R.; Leger, J.; Lee, G. Interaction of tau with the neural
[46] Luo, Y.; Vali, S.; Sun, S.; Chen, X.; Liang, X.; Drozhzhina, T.; plasma membrane mediated by tau's amino-terminal projection
opugaeva, E.; Bezprozvanny, I. Abeta42-Binding peptoids as amy- domain. J. Cell. Biol., 1995, 131, 1327-1340.
loid aggregation inhibitors and detection ligands. ACS Chem. Neu- [66] Spillantini, M. G. Tauopathies: a single therapy for all? Neurobiol.
rosci., 2013, 4, 952-962. Aging, 2012, 3, S34.
[47] Kwiatkowski, D.J.; Mehl, R.; Yin, H.L. Genomic organization and [67] Sergeant, N.; Bretteville, A.; Hamdane, M.; Caillet-Boudin, M.L.;
biosynthesis of secreted and cytoplasmic forms of gelsolin. J. Cell. Grognet, P.; Bombois, S.; Blum, D.; Delacourte, A.; Pasquier, F.;
Biol., 1988, 106, 375-384. Vanmechelen, E.; Schraen-Maschke, S.; Buée, L. Biochemistry of
[48] Chauhan, V.; Ji, L.; Chauhan, A. Anti-amyloidogenic, anti-oxidant Tau in Alzheimer's disease and related neurological disorders. Exp.
and antiapoptotic role of gelsolin in Alzheimer’s disease. Biogeron- Rev. Proteom., 2008, 5, 207-224.
tology, 2008, 9, 381-389. [68] Ferrer, I.; Gomez-Isla, T.; Puig, B.; Freixes, M.; Ribe, E.; Dalfo,
E.; Avila, J. Current advances on different kinases involved in tau
Exploration of Various Proteins for the Treatment of Alzheimer’s Disease Current Drug Metabolism, 2017, Vol. 18, No. 9 813

phosphorylation, and implications in Alzheimer's disease and phagy and attenuates tauopathy in vitro and in vivo. Autophagy,
tauopathies. Curr. Alzheimer Res., 2005, 2, 3-18. 2012, 8, 609-622.
[69] Mazanetz, M.P.; Fischer, P.M. Untangling tau hyperphosphoryla- [81] Necula, M.; Breydo, L.; Milton, S.; Kayed, R.; van der Veer, W.
tion in drug design for neurodegenerative diseases. Nat. Rev. Drug E.; Tone, P.; Glabe, C.G. Methylene blue inhibits amyloid Abeta
Discov., 2007, 6, 464-479. oligomerization by promoting fibrillization. Biochemistry, 2007,
[70] Himmelstein, D.S.; Ward, S.M.; Lancia, J.K.; Patterson, K.R.; 46, 8850-8860.
Binder, L.I. Tau as a therapeutic target in neurodegenerative dis- [82] Pfaffendorf, M.; Bruning, T.A.; Batnik, H.D.; van Zwieten, P.A.
ease. Pharmacol. Ther., 2012, 136, 8-22. The interaction between methylene blue and the cholinergic sys-
[71] Churcher, I. Tau therapeutic strategies for the treatment of Alz- tem. Br. J. Pharmacol., 1997, 122, 95-98.
heimer's disease. Curr. Top. Med. Chem., 2006, 6, 579-595. [83] Atamna, H.; Nguyen, A.; Schultz, C.; Boyle, K.; Newberry, J.;
[72] Wang, J.Z.; Grundke-Iqbal, I.; Iqbal, K. Kinases and phosphatases Kato, H.; Ames, B.N. Methylene blue delays cellular senescence
and tau sites involved in Alzheimer neurofibrillary degeneration. and enhances key mitochondrial biochemical pathways. FASEB J.,
Eur. J. Neurosci., 2007, 25, 59-68. 2008, 22, 703-712.
[73] Gong, C.X.; Iqbal, K. Hyperphosphorylation of microtubule- [84] Daccache, A.; Lion, C.; Sibille, N.; Gerard, M.; Slomianny, C.;
associated protein tau: A promising therapeutic target for Alz- Lippens, G.; Cotelle, P. Oleuropein and derivatives from olives as
heimer disease. Curr. Med. Chem., 2008, 15, 2321-2328. Tau aggregation inhibitors. Neurochem. Int., 2011, 58, 700-707.
[74] Chohan, M.O.; Khatoon, S.; Iqbal, I.G.; Iqbal, K. Involvement of [85] Dickey, C.A.; Kamal, A.; Lundgren, K.; Klosak, N.; Bailey, R.M.;
I2PP2A in the abnormal hyperphosphorylation of tau and its rever- Dunmore, J.; Ash, P.; Shoraka, S.; Zlatkovic, J.; Eckman, C.B.;
sal by memantine. FEBS Lett., 2006, 589, 3973-3979. Patterson, C.; Dickson, D. W.; Nahman, N. S. Jr.; Hutton, M.; Bur-
[75] Kickstein, E.; Krauss, S.; Thornhill, P.; Rutschow, D.; Zeller, R.; rows, F.; Petrucelli, L. The high-affinity HSP90-CHIP complex
Sharkey, J.; Williamson, R.; Fuchs, M.; Köhler, A.; Glossmann, H.; recognizes and selectively degrades phosphorylated tau client pro-
Schneider, R.; Sutherland, C.; Schweiger, S. Biguanide metformin teins. J. Clin. Invest., 2007, 117, 648-658.
acts on tau phosphorylation via mTOR/protein phosphatase 2A [86] Giommarelli, C.; Zuco, V.; Favini, E.; Pisano, C.; Dal Piaz, F.; De
(PP2A) signaling. Proc. Natl. Acad. Sci. USA, 2010, 107, 21830- Tommasi, N.; Zunino, F. The enhancement of antiproliferative and

tio y
21835. proapoptotic activity of HDAC inhibitors by curcumin is mediated
[76] Andrieux, A.; Salin, P.; Schweitzer, A.; Begou, M.; Pachoud, B.; by Hsp90 inhibition. Cell Mol. Life Sci., 2010, 67, 995-1004.

bu nl
Brun, P.; Gory-Fauré, S.; Kujala, P.; Suaud-Chagny, M. F.; Höfle, [87] Ma, Q.L.; Zuo, X.; Yang, F.; Ubeda, O.J.; Gant, D.J.; Alaverdyan,

n
G.; Job, D. Microtubule stabilizer ameliorates synaptic function M.; Teng, E.; Hu, S.; Chen, P.P.; Maiti, P.; Teter, B.; Cole, G. M.;

tri O
and behavior in a mouse model for schizophrenia. Biol. Psychiat., Frautschy, S. A. Curcumin suppresses soluble tau dimers and cor-
2006, 60, 1224-1230. rects molecular chaperone, synaptic, and behavioral deficits in aged
[77] Gozes, I.; Divinski, I.; Piltzer, I. NAP and D-SAL: neuroprotection human tau transgenic mice. J. Biol. Chem., 2013, 288, 4056-4065.
against the beta amyloid peptide (1-42). BMC Neurosci., 2008, 9 [88] Rosenmann, H. Immunotherapy for targeting tau pathology in
is e
(Suppl. 3), S3. Alzheimer’s disease and tauopathies. Curr. Alzheimer Res., 2013,
[78] Gozes, I. NAP (davunetide) provides functional and structural 10, 217-228.
s
neuroprotection. Curr. Pharm. Des., 2011, 17, 1040-1044. [89] Boutajangout, A.; Ingadottir, J.; Davies, P.; Sigurdsson, E.M. Pas-
rD U

[79] Rojo, L.E.; Alzate-Morales, J.; Saavedra, I.N.; Davies, P.; Mac- sive immunization targeting pathological phospho-tau protein in a
cioni, R.B. Selective interaction of lansoprazole and astemizole mouse model reduces functional decline and clears tau aggregates
with tau polymers: potential new clinical use in diagnosis of Alz- from the brain. J. Neurochem., 2011, 118, 658-667.
heimer’s disease. J. Alzheimers Dis., 2010, 19, 573-589. [90] Chai, X.; Wu, S.; Murray, T.K.; Kinley, R.; Cella, C.V.; Sims, H.;
Fo al

[80] Congdon, E.E.; Wu, J.W.; Myeku, N.; Figueroa, Y.H.; Herman, Buckner, N.; Hanmer, J.; Davies, P.; O'Neill, M.J.; Hutton, M.L.;
M.; Marinec, P.S.; Gestwicki, J.E.; Dickey, C.A.; Yu, W.H.; Duff, Citron, M. Passive immunization with anti-Tau antibodies in two
ot on

K.E. Methylthioninium chloride (methylene blue) induces auto- transgenic models: Reduction of Tau pathology and delay of dis-
ease progression. J. Biol. Chem., 2011, 286, 34457-34467.
N rs
Pe

View publication stats

You might also like