ArticlePDF AvailableLiterature Review

Molecular Pathogenesis of Alzheimer's Disease: An Update

Authors:

Abstract and Figures

Dementia is a chronic or progressive syndrome, characterized by impaired cognitive capacity beyond what could be considered a consequence of normal aging. It affects the memory, thinking process, orientation, comprehension, calculation, learning ability, language, and judgment; although awareness is usually unaffected. Alzheimer’s disease (AD) is the most common form of dementia; symptoms include memory loss, difficulty solving problems, disorientation in time and space, among others. The disease was first described in 1906 at a conference in Tubingen, Germany by Alois Alzheimer. One hundred and ten years since its first documentation, many aspects of the pathophysiology of AD have been discovered and understood, however gaps of knowledge continue to exist. This literature review summarizes the main underlying neurobiological mechanisms in AD, including the theory with emphasis on amyloid peptide, cholinergic hypothesis, glutamatergic neurotransmission, the role of tau protein, and the involvement of oxidative stress and calcium.
Content may be subject to copyright.
E-Mail karger@karger.com
Review
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
Molecular Pathogenesis of
Alzheimer’s Disease: An Update
Alfredo Sanabria-Castro Ileana Alvarado-Echeverría Cecilia Monge-Bonilla
Research Unit, Hospital San Juan de Dios, Costa Rican Social Security Fund (CCSS), San José , Costa Rica
Search Strategy and Selection Criteria
A literature review was conducted in January 2016 us-
ing PubMed, Ovid, and Science Direct with the following
descriptors: Alzheimer’s disease (AD), neurobiology, pa-
thology, review. Results obtained ranged between 150
and 2,200 records after the combination of different key-
words. Scientific publications between 1996 and 2015
either in English or Spanish, discussing the neuromolecu-
lar hypotheses of AD were selected. Papers concerning
clinical presentation, diagnostic methods, and treatment
were excluded.
Alzheimer’s disease (AD) was first described in 1906
at a conference in Tubingen, Germany by Alois Alzheimer
[1] , as a “peculiar severe disease process of the cerebral
cortex.” In recent times, AD is considered a chronic or
progressive syndrome, characterized by impaired cogni-
tive capacity beyond what could be considered a conse-
quence of normal aging; that affects the memory, think-
ing, orientation, comprehension, learning, language, and
judgment. More than one hundred years have passed
since the first pathophysiological aspects of AD were de-
scribed. Neurobiological mechanisms underlying AD
have been a key element in the understanding of the pa-
thology, currently the most important alterations identi-
Keywords
Alzheimer’s disease · Dementia · Amyloid · Neurobiology ·
Pathogenesis · Review
Abstract
Dementia is a chronic or progressive syndrome, character-
ized by impaired cognitive capacity beyond what could be
considered a consequence of normal aging. It affects the
memory, thinking process, orientation, comprehension, cal-
culation, learning ability, language, and judgment; although
awareness is usually unaffected. Alzheimer’s disease (AD) is
the most common form of dementia; symptoms include
memory loss, difficulty solving problems, disorientation in
time and space, among others. The disease was first de-
scribed in 1906 at a conference in Tubingen, Germany by
Alois Alzheimer. One hundred and ten years since its first
documentation, many aspects of the pathophysiology of AD
have been discovered and understood, however gaps of
knowledge continue to exist. This literature review summa-
rizes the main underlying neurobiological mechanisms in
AD, including the theory with emphasis on amyloid peptide,
cholinergic hypothesis, glutamatergic neurotransmission,
the role of tau protein, and the involvement of oxidative
stress and calcium. © 2017 S. Karger AG, Basel
Received: July 16, 2016
Accepted: July 26, 2016
Published online: April 21, 2017
Alfredo Sanabria-Castro
Research Unit, Hospital San Juan de Dios
Costa Rican Social Security Fund (CCSS)
Paseo Colón, Streets 14–20 San José 10103 (Costa Rica)
E-Mail asccheo @ yahoo.com
© 2017 S. Karger AG, Basel
www.karger.com/aon
Molecular Pathogenesis of AD:
An Update
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
47
fied can be explained through: the amyloid peptide theo-
ry, the cholinergic hypothesis that includes glutamatergic
neurotransmission alterations, the role of tau protein,
and the involvement of oxidative stress (OS) and calcium.
The Cholinergic Hypothesis
At a molecular level, the cholinergic hypothesis is the
first and most studied approach that describes AD patho-
physiology. It was defined more than 30 years ago as a
primary degenerative process capable of selectively dam-
aging groups of cholinergic neurons in the hippocampus,
frontal cortex, amygdala, nucleus basalis, and medial sep-
tum, regions and structures that serve important func-
tional roles in conscious awareness, attention, learning,
memory, and other mnemonic processes
[2] . This selec-
tive alteration generates a downregulation of cholinergic
markers such as acetyltransferase and acetylcholinester-
ase that associate with the onset of cognitive impairment
[3, 4] ; through the existence of proportionality between
the decrease in cholinergic markers, the density of neuro-
fibrillary alterations, and the severity of the pathology.
The main findings supporting this premise are the fact
that non-selective muscarinic antagonists such as sco-
polamine-induced cognitive impairment, favor the pro-
duction of beta-amyloid peptide and decrease the activ-
ity of α-secretase
[5] . Some triterpenoid saponins have
shown to reduce scopolamine-induced amnesia
[6–8]
and non-selective and selective muscarinic agonists have
shown to improve learning and memory. Selective M1
muscarinic agonists are a pivotal target that link major
hallmarks of AD. The exact molecular mechanisms of the
effect of cholinergic drugs in learning and memory, and
their clinical treatment viability are still being studied
[9–
12] .
This hypothesis was reinforced through immunohis-
tochemical, neuroimaging, and other analyses that re-
vealed: a decrease in the number and density of nicotinic
receptors in AD patients (mainly α4β2 subtype), a re-
duced expression of α3, α4, and α7 subunits at cortex and
hippocampus, and a decline in the binding ability of α7
hippocampal and α4 cortical receptors
[13–15] .
The main alterations in cholinergic neurons consid-
ered in this hypothesis are: choline uptake, impaired ace-
tylcholine release, deficits in the expression of nicotinic
and muscarinic receptors, dysfunctional neurotrophin
support, and deficits in axonal transport
[16–18] . Recent
studies have shown that amyloid β interacts with cholin-
ergic receptors affecting their function
[19] .
Because the cholinergic and glutamatergic systems sig-
nificantly interact during neurotransmission alterations
in the glutamatergic, signaling has been associated with
cholinergic disruptions found in AD
[20] , an aspect that
enhances cholinergic hypothesis. This postulate dictates
that acetylcholine and its receptors, especially (α7)
5 are
considered as neuroprotective by modulating glutamate-
mediated neuronal excitability
[21, 22] . In AD abnormal-
ities in glutamatergic, neurotransmission is initially ob-
served at the entorhinal cortex (EC), which is followed by
further neurotransmission defects in the hippocampus,
amygdala, frontal cortex, and parietal cortex
[23] .
The physiological glutamatergic neurotransmission in
the hippocampus produces a cytosolic calcium signal,
which mediates synaptic plasticity phenomena such as
long-term potentiation (LTP), encouraging learning and
memory consolidation
[24] . However, a sustained in-
crease in calcium, sodium, and chloride ions as a result of
the hyperactivation of NMDA glutamate receptors has
been associated with excessive depolarization of the post-
synaptic membrane, onset neurodegenerative processes
and cell death
[25–27] . Likewise, an increase in intraneu-
ronal calcium as a consequence of a dysfunctional gluta-
matergic neurotransmission can generate a long-lasting
depression in the cerebellum (LTD), with calcium over-
load in mitochondria, activation of nitric oxide synthesis,
generation of free radicals, OS, initiation apoptosis and
neuronal death
[28–31] .
Experimentally incubating neurons with glutamate
promotes the deposit of filaments similar to neurofibril-
lary tangles observed in AD. Also, the exposure of neuro-
nal cultures to amyloid β promotes glutamate-induced
neurotoxicity and regulates the expression of NMDA
receptors on the membrane
[32, 33] .
At the synaptic level, the lack of enzymes responsible
for the degradation of glutamate causes neuronal trans-
porters at the neuronal and glial levels to be responsible
for the reuptake of the excess of neurotransmitter
[34] .
In AD, the inhibition of presynaptic and glial glutamate
transport
[35] , the reduced activity of glutamine synthe-
tase (converts glutamate to glutamine)
[36] , the discrete
depolarization of neurons, and stimulation of nitric ox-
ide production by amyloid β
[23] favor the prolonged
presence of extra neuronal glutamate, and hence the
continued receptor stimulation and excitotoxicity
[16,
34] .
The cholinergic hypothesis has served as a basis for the
majority of treatment strategies and drug development
approaches (acetylcholinesterase inhibitors, cholinergic
precursors, cholinergic receptor agonists, allosteric cho-
Sanabria-Castro/Alvarado-Echeverría/
Monge-Bonilla
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
48
linergic receptor potentiators, NMDA receptor blockers)
for AD. Currently there is consensus that the observed
relationship between cognitive impairment and de-
creased cholinergic transmission in the brain plays an im-
portant role in AD but by itself does not establish defini-
tive causation of the disease
[37, 38] .
The Amyloid Hypothesis
In the amyloid cascade hypothesis of AD, the disease
is analyzed as a series of abnormalities in the process and
secretion of the amyloid precursor protein (APP), where
an inequality between production and clearance of amy-
loid β is the triggering event and the most important fac-
tor; responsible for other abnormalities observed in AD
[39] . Amyloid β is a peptide with high resistance to pro-
teolytic degradation. It consists of 37–43 amino acids, in
which the isoforms 1–40 and 1–42 are the most common
[40] . The 1–42 amyloid peptide isoform is the most hy-
drophobic and is considered to have the greatest toxicity
[23] . Because of its physical characteristics, it often ac-
quires the configuration of a β-pleated sheet
[41] , show-
ing a greater tendency to aggregate and form the core of
the amyloid plaque
[42–44] . It is the main component of
amyloid neuritic plaques
[43, 45] .
The processing of APP at the plasma membrane con-
stitutes the origin of the amyloid peptide ( Fig.1 ). APP is
Abnormal Normal
APPsį
APP
APPsDŽ
Oligomer
DŽƩ
Amyloid
plaque
DŽ
DŽ$5
*35
&;&5
Alterations
T phosphorilation
Calcium dysfunction
Mitochondrial
dysfunction
Synaptic alteration
Cognitive impairment
'25
$$5
P$&K5
P*OX5*, 5
+7$&
&5+5
3$&5
FGF, EGF, NGF
Cytokines
Hormones
ADAM
BACE1
C99 & AICD
AICD
YY
S
į
DŽƩ
Fig. 1. Amyloidogenic and non-amyloidogenic processing of the
APP. The top part of the figure shows the 2 main paths in the
processing of APP, the most important elements involved, and
the main alterations associated with the amyloidogenic pathway
of APP. The bottom part of the figure shows potential modula-
tors of the secretase activity (activation of receptors, growth fac-
tors, cytokines, hormones). α, α-secretase activity; β, β-secretase
activity; γ, γ-secretase activity; BACE1, β-site – APP – cleaving
enzyme 1; βA, amyloid β peptide; ADAM, α-secretase; APPsα
and APPsβ, soluble portions produced by the effect of α- and
β-secretase; C83, fragment of 83 amino acids of the carboxyl ter-
minal portion produced by the effect of α-secretase; C99, frag-
ment of 99 amino acids of the carboxyl terminal portion pro-
duced by effect of β-secretase; p3, peptide resulting from
γ-secretase; AICD, carboxy-terminal fragment referred to as the
intracellular domain of APP; T, Tau protein; mAChRs, musca-
rinic acetylcholine receptors; mGluR, metabotropic glutamate
receptors; 5HT, serotonin receptors; CRHR1, receptor 1 of the
corticotropin-releasing hormone; PAC1R, receptor 1 of pituitary
adenylate cyclase; FGF, EGF, NGF, growth factors of fibroblasts,
epidermis and the nervous system respectively; DOR, opioid
receptor δ; A2AR, adenosine receptor 2A; β2-AR, β2 adrenergic
receptor; GPR3, receptor 3 coupled to protein G; CXCR2,
chemokine receptor 2.
Molecular Pathogenesis of AD:
An Update
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
49
a transmembrane glycoprotein of type I and its specific
function is unclear; however it is known that its expres-
sion is increased during cellular stress phenomena. APP
processing is performed by a series of ruptures, which in-
volves an initial breakdown by enzymes with α-secretase
activity, mainly enzymes belonging to the disintegrin and
metalloprotease (ADAM) family including: ADAM10,
ADAM17, and ADAM19
[45, 46] . ADAM activity can be
modulated by different situations such as receptor activa-
tion, growth factors, cytokines, and hormones. The exci-
sion produced by α-secretase leads to the formation and
release of a peptide of the amino terminal portion called
APPs α, which is soluble under certain conditions
[47]
and a fragment of the carboxyl terminal portion (C83)
[48] . The APPsα is found in lower quantities in AD pa-
tients
[49] and has been associated with trophic and neu-
roprotective functions
[50] .
In patients with AD, the first APP rupture at an extra-
cellular level, generates a shorter soluble amino terminal
portion (APPsβ) and a longer terminal carboxyl fragment
(C99)
[51, 52] . This split is performed mainly by BACE1
(β- site – APP – cleaving enzyme), an ubiquitous trans-
membrane protease with β-secretase activity. BACE1 ex-
pression can be modulated by frequently seen situations
in neurodegenerative diseases and aging such as OS, isch-
emia, inflammation, hypoxia, and trauma
[51, 53–55] .
After γ-secretase, consisting of a heteromeric complex
of 4 subunits called: presenilins (PSEN1 and PSEN2),
nicastrin (NCSTN), APH-1, from the acronym anterior
pharynx defensive phenotype 1 (APH-1a and APH-1b)
and PEN – 2 (PS-enhancer-2)
[56] produces a cut in the
γ site releasing the carboxyl terminal fragment named
APP intracellular domain (AICD) and producing the am-
yloid β peptide, which is secreted, aggregated, and accu-
mulated in extracellular plaques due to its low solubility
( Fig.2 ) [57] . It has been observed that APH-1 inhibits the
production of amyloid β peptide while the PEN-2 favors
it
[58, 59] .
Several studies have shown the in vitro and in vivo
neurotoxicities of various forms of amyloid β
[60] ; how-
ever, the exact mechanisms are quite complex and not
fully understood
[37, 52] . So far the relation between the
specific site of action of amyloid β and its structural di-
versity is unknown; what is clear is that the amino acid
sequence that is contained between positions 25 and 35
of the primary structure has greater neurotoxicity.
Concentrations of amyloid β peptide are determined
by the balance between generation and clearance; in AD
APP
1
4
40
42
5
C31
Nicastrin
Y-Secretase
Oligomerization
BACE PEN-2 Presenilin
AICD
2b
2a
APH-1
Y
a
ı
3
DŽ6HFUHWDVH
Fig. 2. Production of amyloid β peptide from sequential proteolytic
breaks of the APP. The figure shows the sequence of phases in the
processing of APP in the formation of amyloid β peptide and its
oligomerization. Numbers (1, 2a, 2b, 3, 4, and 5) refer to intervention
sites where the production of amyloid β peptide could be modulated
with possible therapeutic utility. α, α-secretase cut site; γ, γ-secretase
cut site γ; ε, cut site ε of γ-secretase; BACE, β-secretase; Aβ, amyloid
β peptide; AICD, carboxy-terminal fragment referred to as APP in-
tracellular domain.
Sanabria-Castro/Alvarado-Echeverría/
Monge-Bonilla
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
50
patients, a clearance abnormality leads to the accumula-
tion of amyloid β in the brain
[61] . The transport of amy-
loid β through the blood-brain barrier is mediated by re-
ceptors, the passage of the peptide from the brain to the
blood occurs by interaction with LRP-1 receptors (Lipo-
protein receptor-related protein-1) and the action of p-
glycoproteins. Increased levels of amyloid β and aging,
decrease of the expression of LRP-1 receptors results in
an accumulation of amyloid peptide in the central ner-
vous system (CNS).
The transit of amyloid β peptide into the brain through
the blood-brain barrier
[62] also occurs via receptors, pri-
marily through multi-ligand AGE (advanced glycation
end products) receptors or RAGE. RAGE expression is
determined by the concentration of its own ligands. In
contrast to the expression decrease of LPR-1s due to high
levels of amyloid β in the brain, RAGE increases the ex-
pression under these conditions.
Since RAGE interaction with amyloid β causes: in-
flammatory responses at the endothelium level, endothe-
lial cell apoptosis, decreased cerebral blood flow, and
suppression of LTP; RAGE may play a role in the devel-
opment of neurovascular changes observed in AD
[62–
65] .
Proteolytic degradation of amyloid β peptide is car-
ried out mainly by neprilysin (NEP) and the insulin-
degrading enzyme (IDE). During aging and in AD pa-
tients, the expression of NEP and IDE decreases, caus-
ing an increase in the concentration of amyloid β peptide
in the brain
[61, 66] . In patients with AD, decreased
quantity and activity of NEP is seen especially in the
cortex and hippocampus, but not in other regions of the
brain.
The secretion of IDE in the brain is regulated by the
microglia, and similar to NEP its distribution and con-
centration also presents differences in AD patients. Low-
er concentrations have been identified in the cortex and
hippocampus, where the predominant form has a higher
degree of oxidation
[9] .
The endosomal lysosomal pathway is also an impor-
tant regulator of the processing of APP
[67, 68] and of
the tau protein metabolism
[69] . Because of the impor-
tance of this pathway for cellular maintenance and its
role in the immune system, recent studies suggest that
dysfunctions in neuronal autophagy causing an in-
crease in the amount and size of endosomes at the cel-
lular level, may be involved in the pathogenesis of AD
[70–74] , as these changes are seen before the appear-
ance of senile plaques and neurofibrillary tangles in the
brain
[72, 74] . Other studies report the absence of a di-
rect relationship between quantity and size of lyso-
somes and AD [75] .
The amyloid cascade hypothesis has the highest accep-
tance rate; however, current studies support the idea that
not only amyloid β but other fragments from the process-
ing of APP, such as C83 or AICD, contribute to the patho-
genesis of AD resulting in difficulty to attribute the path-
ological features of the disease exclusively to the amyloid
peptide
[60] .
The Tau (τ) Protein
The amyloid cascade hypothesis consists of the pro-
duction and accumulation of amyloid β peptide as the
beginning of the disease process; however, it does not
completely explain the etiopathogenesis of AD. In this
hypothesis, the τ protein arises as a secondary patho-
genic event that subsequently causes neurodegeneration
[76, 77] . In vitro experiments using various cell types,
ranging from neuronal cell lines to primary hippocam-
pal and cortical neurons and hippocampal organotypic
cultures, have demonstrated that amyloid β induces
τ-alterations
[78–80] . These include mainly an increased
phosphorylation and cytoplasmic and dendritic translo-
cation often linked to neurodegeneration
[81] .
The τ protein is a highly soluble protein that re-
latesto the microtubules and its function under normal
conditions consists of stabilizing them. These microtu-
bules provide support for structural changes, axonal
transport, and neuronal growth
[69, 82, 83] . In the
CNS, the τ protein presents itself in 6 different isoforms
that vary in the number of binding sites for microtu-
bules and the amount of exons they possess
[82] . In AD,
dysfunctions occur in phosphorylation processes of τ
protein, resulting in a hyperphosphorylation of the
molecule.
Hyperphosphorylated protein τ presents aberrant ag-
gregation with the cytoskeletal proteins; it shows a lower
grade of interaction with microtubules which favors an
increase of free tau protein that leads to greater aggrega-
tion and fibrillization of itself, with the consequent mal-
function of axonal transport
[84, 85] .
Scientific literature reports changes in τ protein and
amyloid β oligomers as the most important factors re-
sponsible for neuronal dysfunction in the pathogenesis of
AD
[46, 86] . Likewise neurofibrillary tangles observed
initially in the EC and hippocampus subsequently extend
to the amygdala and cortical areas (temporal, frontal, and
parietal)
[57, 85] .
Molecular Pathogenesis of AD:
An Update
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
51
Contribution of OS in the Pathogenesis of AD
It has been long recognized that one of the common
characteristics in neurodegenerative diseases is the rela-
tionship between OS and neuronal apoptosis
[87] . OS is
a condition in which the balance between production of
active reactive oxygen species (ROS) and the level of an-
tioxidants is significantly disturbed, resulting in cell dam-
age. ROS chemically interact with biological molecules
such as nucleic acids, proteins and lipids, and cell organ-
elles
[88] .
In addition to the established pathology of senile
plaques and neurofibrillary tangles, the presence of ex-
tensive OS is a characteristic of AD brains. The accumu-
lation of free radical damage, alterations in the activities
or expression of antioxidant enzymes such as superoxide
dismutase and catalase are also present in AD patients
[89] . Although OS is an important factor in AD patho-
genesis, the mechanisms by which the redox balance is
altered and the sources of free radicals are not exactly
known. It has been demonstrated that abnormal accumu-
lation of amyloid β is capable of promoting the formation
of ROS through a mechanism that involves the activation
of NMDA receptors
[90] , and that OS may augment am-
yloid β production and aggregation as well facilitate tau
phosphorylation and polymerization, forming a vicious
cycle that promotes the initiation and progression of AD
[91] .
Neuronal mitochondria (essential for cellular
metabolism) show metabolic abnormalities in AD
models. It has been demonstrated that mitochondria
are quite vulnerable to OS, which may directly disrupt
its functions (energy production, decrease of antioxi-
dant enzymes, and loss of membrane potential), gener-
ating a further increase in ROS levels that finally pro-
duce cell death by caspase activation and apoptosis
[92–
94] .
Calcium Homeostasis in AD
Calcium, an ubiquitous intracellular messenger, regu-
lates multiple physiological functions, generating con-
centration gradients and binding to several proteins, re-
ceptors, and ion channels. The regulation of intracellular
calcium homeostasis is a very complex mechanism that is
vital for several cellular pathways and is thus involved in
cell survival and death. Two organelles play a major role
in calcium homeostasis, the endoplasmic reticulum (ER)
and mitochondria, whereas ATPase calcium pump and
the sodium-calcium exchanger are the 2 main systems in-
volved in calcium efflux through the plasma membrane
[95, 96] . Ca 2+ is continuously exchanged between the cy-
tosol and the lumen of the ER. Overload of intracellular
calcium due to a blockage or dysfunction of the transport
system leads to the cleavage of several proteins and other
substrates, OS, perturbs energy production
[97] , stimu-
lates protein production (amyloid β and τ protein)
[98],
and induces cell death through necrosis and/or apoptosis
[99, 100] .
In AD, the ability of neurons to regulate the influx,
efflux, and subcellular compartmentalization of calcium
is compromised
[101] . These disruptions involve sev-
eral mechanisms, such as alterations of calcium buffer-
ing capacities, deregulation of calcium channel activi-
ties, excitotoxicity or disruption of mitochondrial func-
tions. Alterations resulting from calcium disruption are
the result of age-related OS, metabolic impairment in
combination with disease-related accumulation of Aβ
oligomers and the presence of mutations of genes that
encode presenilin
[102] . Particularly, Aβ may promote
cellular calcium overload by inducing membrane-asso-
ciated OS and forming pores in the membrane
[103–
105] .
Conclusion
During the last decades, advances in cellular biology
have been essential for understanding the molecular
mechanisms underlying AD. Currently it is known that
the molecular pathogenesis of AD is complex and in-
volves several theories or hypothesis where many diverse
factors interrelate. However, none of these postulates
alone is able to clarify the entire aspect regarding the pa-
thology, and further studies are required. Aspects like the
initial causes of the disease such as the abnormal forma-
tion of amyloid β, and the mechanisms by which it affects
neurons and nicotinic acetylcholine receptors and the re-
lation between the disruption of cholinergic pathways
and the cognitive deficits of AD are still not fully under-
stood.
New discoveries that contribute to the elucidation of
the molecular pathogenesis of AD and its relations are
crucial because they can allow the development of new
therapeutic strategies for the treatment of a condition
where current drug therapy lacks the ability to prevent its
occurrence and progression.
Sanabria-Castro/Alvarado-Echeverría/
Monge-Bonilla
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
52
1 Alzheimer A: Über einen eigenartigen
schweren erkrankungsprozeβ der hirnrincle.
Neurol Cent 1906;
25: 1134.
2 Terry AV Jr, Buccafusco JJ: The cholinergic
hypothesis of age and Alzheimer’s disease-re-
lated cognitive deficits: recent challenges and
their implications for novel drug develop-
ment. J Pharmacol Exp Ther 2003;
306: 821–
827.
3 Schaeffer EL, Gattaz WF: Cholinergic and
glutamatergic alterations beginning at the
early stages of Alzheimer disease: participa-
tion of the phospholipase A2 enzyme. Psy-
chopharmacology (Berl) 2008;
198: 1–27.
4 Watanabe S, Kato I, Koizuka I: Retrograde-la-
beling of pretecto-vestibular pathways in cats.
Auris Nasus Larynx 2003;
30(suppl):S35–S40.
5 Liskowsky W, Schliebs R: Muscarinic acetyl-
choline receptor inhibition in transgenic Al-
zheimer-like Tg2576 mice by scopolamine fa-
vours the amyloidogenic route of processing
of amyloid precursor protein. Int J Dev Neu-
rosci 2006;
24: 149–156.
6 Li Y, Yuan X, Shen Y, Zhao J, Yue R, Liu F, et
al: Bacopaside I ameliorates cognitive impair-
ment in APP/PS1 mice via immune-mediated
clearance of β-amyloid. Aging (Albany NY)
2016;
8: 521–533.
7 Saraf MK, Anand A, Prabhakar S: Scopol-
amine induced amnesia is reversed by Bacopa
monniera through participation of kinase-
CREB pathway. Neurochem Res 2010;
35:
279–287.
8 Saraf MK, Prabhakar S, Khanduja KL, Anand
A: Bacopa monniera attenuates scopolamine-
induced impairment of spatial memory in
mice. Evid Based Complement Alternat Med
2011;
2011: 236186.
9 Caccamo A, Oddo S, Billings LM, Green KN,
Martinez-Coria H, Fisher A, et al: M1 recep-
tors play a central role in modulating AD-like
pathology in transgenic mice. Neuron 2006;
49: 671–682.
10 Fisher A: M1 muscarinic agonists target ma-
jor hallmarks of Alzheimer’s disease – an up-
date. Curr Alzheimer Res 2007;
4: 577–580.
11 Fisher A, Medeiros R, Barner N, Natan N,
Brandeis R, Elkon H, et al: M1 muscarinic ag-
onists and a multipotent activator of sigma1/
M1 muscarinic receptors: future therapeutics
of Alzheimer’s disease. Alzheimers Demen
2014;
10:P123.
12 Fisher A: M1 muscarinic agonists target ma-
jor hallmarks of Alzheimer’s disease – the piv-
otal role of brain M1 receptors. Neurodegener
Dis 2008;
5: 237–240.
13 Ellis JR, Ellis KA, Bartholomeusz CF, Harri-
son BJ, Wesnes KA, Erskine FF, et al: Musca-
rinic and nicotinic receptors synergistically
modulate working memory and attention in
humans. Int J Neuropsychopharmacol 2006;
9: 175–189.
14 Ringman JM, Cummings JL: Current and
emerging pharmacological treatment options
for dementia. Behav Neurol 2006;
17: 5–16.
15 Wu J, Ishikawa M, Zhang J, Hashimoto K:
Brain imaging of nicotinic receptors in Al-
zheimer’s disease. Int J Alzheimers Dis 2010;
2010: 548913.
16 Wenk GL: Neuropathologic changes in Al-
zheimer’s disease: potential targets for treat-
ment. J Clin Psychiatry 2006;
67(suppl 3):3–7;
quiz 23.
17 Mesulam M: The cholinergic lesion of Al-
zheimer’s disease: pivotal factor or side show?
Learn Mem 2004;
11: 43–49.
18 Shen J, Wu J: Nicotinic cholinergic mecha-
nisms in Alzheimer’s disease. Int Rev Neuro-
biol 2015;
124: 275–292.
19 Ni R, Marutle A, Nordberg A: Modulation of
α7 nicotinic acetylcholine receptor and fibril-
lar amyloid-β interactions in Alzheimer’s dis-
ease brain. J Alzheimers Dis 2013;
33: 841–
851.
20 Dong XX, Wang Y, Qin Z: Molecular mech-
anisms of excitotoxicity and their rele-
vanceto pathogenesis of neurodegenerative
diseases. Acta Pharmacol Sin 2009;
30: 379–
387.
21 Lin H, Vicini S, Hsu FC, Doshi S, Takano H,
Coulter DA, et al: Axonal α7 nicotinic ACh
receptors modulate presynaptic NMDA re-
ceptor expression and structural plasticity of
glutamatergic presynaptic boutons. Proc Natl
Acad Sci U S A 2010;
107: 16661–16666.
22 Geerts H, Grossberg GT: Pharmacology of
acetylcholinesterase inhibitors and N-meth-
yl-D-aspartate receptors for combination
therapy in the treatment of Alzheimer’s dis-
ease. J Clin Pharmacol 2006;
46(7 suppl 1):8S–
16S.
23 Mohandas E, Rajmohan V, Raghunath B:
Neurobiology of Alzheimer’s disease. Indian
J Psychiatry 2009;
51: 55.
24 Lynch MA: Long-term potentiation and
memory. Physiol Rev 2004;
84: 87–136.
25 Beck J, Lenart B, Kintner DB, Sun D: Na-K-Cl
cotransporter contributes to glutamate-medi-
ated excitotoxicity. J Neurosci 2003;
23: 5061–
5068.
26 Friedman LK: Calcium: a role for neuropro-
tection and sustained adaptation. Mol Interv
2006;
6: 315–329.
27 Szado T, Vanderheyden V, Parys JB, De
Smedt H, Rietdorf K, Kotelevets L, et al: Phos-
phorylation of inositol 1,4,5-trisphosphate re-
ceptors by protein kinase B/Akt inhibits Ca2+
release and apoptosis. Proc Natl Acad Sci
USA 2008;
105: 2427–2432.
28 Fan MMY, Raymond LA: N-methyl-D-aspar-
tate (NMDA) receptor function and excito-
toxicity in Huntington’s disease. Prog Neuro-
biol 2007;
81: 272–293.
29 Navarro A, Boveris A: The mitochondrial en-
ergy transduction system and the aging pro-
cess. Am J Physiol Cell Physiol 2007;
292:
C670–C686.
30 Jung KH, Chu K, Lee ST, Park HK, Kim JH,
Kang KM, et al: Augmentation of nitrite ther-
apy in cerebral ischemia by NMDA receptor
inhibition. Biochem Biophys Res Commun
2009;
378: 507–512.
31 Ndountse LT, Chan HM: Role of N-methyl-
D-aspartate receptors in polychlorinated bi-
phenyl mediated neurotoxicity. Toxicol Lett
2009;
184: 50–55.
32 Snyder EM, Nong Y, Almeida CG, Paul S,
Moran T, Choi EY, et al: Regulation of NMDA
receptor trafficking by amyloid-beta. Nat
Neurosci 2005;
8: 1051–1058.
33 Parameshwaran K, Dhanasekaran M, Suppi-
ramaniam V: Amyloid beta peptides and glu-
tamatergic synaptic dysregulation. Exp Neu-
rol 2008;
210: 7–13.
34 Butterfield DA, Pocernich CB: The glutama-
tergic system and Alzheimer’s disease: thera-
peutic implications. CNS Drugs 2003;
17: 641–
652.
35 Doraiswamy PM: Non-cholinergic strategies
for treating and preventing Alzheimer’s dis-
ease. CNS Drugs 2002;
16: 811–824.
36 Walton HS, Dodd PR: Glutamate-glutamine
cycling in Alzheimer’s disease. Neurochem
Int 2007;
50: 1052–1066.
37 Peña F, Gutiérrez-Lerma A, Quiroz-Baez R,
Arias C: The role of beta-amyloid protein in
synaptic function: implications for Alzheim-
er’s disease therapy. Curr Neuropharmacol
2006;
4: 149–163.
38 Doggrell SA, Evans S: Treatment of dementia
with neurotransmission modulation. Expert
Opin Investig Drugs 2003;
12: 1633–1654.
39 Cummings JL, Doody R, Clark C: Disease-
modifying therapies for Alzheimer disease:
challenges to early intervention. Neurology
2007;
69: 1622–1634.
40 Deane R, Bell RD, Sagare A, Zlokovic BV:
Clearance of amyloid-beta peptide across the
blood-brain barrier: implication for therapies
in Alzheimer’s disease. CNS Neurol Disord
Drug Targets 2009;
8: 16–30.
41 Jucker M, Walker LC: Neurodegeneration:
amyloid-β pathology induced in humans. Na-
ture 2015;
525: 193–194.
42 McGowan E, Pickford F, Kim J, Onstead L,
Eriksen J, Yu C, et al: Abeta42 is essential for
parenchymal and vascular amyloid deposi-
tion in mice. Neuron 2005;
47: 191–199.
43 Bertram L, Tanzi RE: Thirty years of Alzheim-
er’s disease genetics: the implications of sys-
tematic meta-analyses. Nat Rev Neurosci
2008;
9: 768–778.
44 Wu LG, Saggau P: Presynaptic inhibition of
elicited neurotransmitter release. Trends
Neurosci 1997;
20: 204–212.
45 Leão RN, Colom LV, Borgius L, Kiehn O,
Fisahn A: Medial septal dysfunction by Aβ-
induced KCNQ channel-block in glutamatergic
neurons. Neurobiol Aging 2012;
33: 2046–2061.
46 De Strooper B: Proteases and proteolysis in
Alzheimer disease: a multifactorial view on the
disease process. Physiol Rev 2010;
90: 465–494.
47 Tanzi RE, Bertram L: Twenty years of the Al-
zheimer’s disease amyloid hypothesis: a ge-
netic perspective. Cell 2005;
120: 545–555.
References
Molecular Pathogenesis of AD:
An Update
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
53
48 Lichtenthaler SF, Haass C: Amyloid at the
cutting edge: activation of alpha-secretase
prevents amyloidogenesis in an Alzheimer
disease mouse model. J Clin Invest 2004;
113:
1384–1387.
49 Sennvik K, Fastbom J, Blomberg M, Wahlund
LO, Winblad B, Benedikz E: Levels of alpha-
and beta-secretase cleaved amyloid precursor
protein in the cerebrospinal fluid of Alzheim-
er’s disease patients. Neurosci Lett 2000;
278:
169–172.
50 Puzzo D, Privitera L, Leznik E, Fà M, Stanisze-
wski A, Palmeri A, et al: Picomolar amyloid-
beta positively modulates synaptic plasticity
and memory in hippocampus. J Neurosci
2008;
28: 14537–14545.
51 Velliquette RA, O’Connor T, Vassar R: En-
ergy inhibition elevates beta-secretase levels
and activity and is potentially amyloidogenic
in APP transgenic mice: possible early events
in Alzheimer’s disease pathogenesis. J Neuro-
sci 2005;
25: 10874–10883.
52 Siegel GS, Chauhan N, Karczmar AG: Links
between Amyloid and Tau Biology in Al-
zheimer’s Disease and Their Cholinergic As-
pects. Exploring the Vertebrate Central Cho-
linergic Nervous System. New York, 2007, pp
597–603.
53 Wen Y, Onyewuchi O, Yang S, Liu R, Simp-
kins JW: Increased beta-secretase activity and
expression in rats following transient cerebral
ischemia. Brain Res 2004;
1009: 1–8.
54 Bourne KZ, Ferrari DC, Lange-Dohna C,
Rossner S, Wood TG, Perez-Polo JR: Differ-
ential regulation of BACE1 promoter activity
by nuclear factor-kappaB in neurons and glia
upon exposure to beta-amyloid peptides. J
Neurosci Res 2007;
85: 1194–1204.
55 Zhang X, Zhou K, Wang R, Cui J, Lipton SA,
Liao FF, et al: Hypoxia-inducible factor 1al-
pha (HIF-1alpha)-mediated hypoxia increas-
es BACE1 expression and beta-amyloid gen-
eration. J Biol Chem 2007;
282: 10873–10880.
56 De Strooper B: Aph-1, Pen-2, and Nicastrin
with Presenilin generate an active gamma-
Secretase complex. Neuron 2003;
38: 9–12.
57 Goedert M, Spillantini MG: A century of Al-
zheimer’s disease. Science 2006;
314: 777–781.
58 Seo SJ, Hwang DY, Cho JS, Chae KR, Kim CK,
Shim SB, et al: PEN-2 overexpression induces
gamma-secretase protein and its activity with
amyloid beta-42 production. Neurochem Res
2007;
32: 1016–1023.
59 Uemura K, Lill CM, Li X, Peters JA, Ivanov A,
Fan Z, et al: Allosteric modulation of PS1/
gamma-secretase conformation correlates
with amyloid beta(42/40) ratio. PLoS One
2009;
4:e7893.
60 Simon A, Frechilla D, del Río J: Perspectivas
sobre la hipótesis de la cascada del amiloide
en la enfermedad de Alzheimer. Rev Neurol
2010;
50: 667–675.
61 Wang DS, Dickson DW, Malter JS: Beta-am-
yloid degradation and Alzheimer’s disease. J
Biomed Biotechnol 2006;
2006: 58406.
62 Chen X, Walker DG, Schmidt AM, Arancio
O, Lue LF, Yan SD: RAGE: a potential target
for Abeta-mediated cellular perturbation in
Alzheimer’s disease. Curr Mol Med 2007;
7:
735–742.
63 McGeer PL, Rogers J, McGeer EG: Inflamma-
tion, anti-inflammatory agents and Alzheim-
er disease: the last 12 years. J Alzheimers Dis
2006;
9(3 suppl):271–276.
64 Origlia N, Righi M, Capsoni S, Cattaneo A,
Fang F, Stern DM, et al: Receptor for ad-
vanced glycation end product-dependent ac-
tivation of p38 mitogen-activated protein ki-
nase contributes to amyloid-beta-mediated
cortical synaptic dysfunction. J Neurosci
2008;
28: 3521–3530.
65 Aisen PS: Alzheimer’s disease therapeutic re-
search: the path forward. Alzheimers Res
Ther 2009;
1: 2.
66 Miners JS, Baig S, Tayler H, Kehoe PG, Love
S: Neprilysin and insulin-degrading enzyme
levels are increased in Alzheimer disease in
relation to disease severity. J Neuropathol Exp
Neurol 2009;
68: 902–914.
67 Grbovic OM, Mathews PM, Jiang Y, Schmidt
SD, Dinakar R, Summers-Terio NB, et al:
Rab5-stimulated up-regulation of the endo-
cytic pathway increases intracellular beta-
cleaved amyloid precursor protein carboxyl-
terminal fragment levels and Abeta produc-
tion. J Biol Chem 2003;
278: 31261–31268.
68 Pasternak SH, Callahan JW, Mahuran DJ: The
role of the endosomal/lysosomal system in
amyloid-beta production and the pathophys-
iology of Alzheimer’s disease: reexamining
the spatial paradox from a lysosomal perspec-
tive. J Alzheimers Dis 2004;
6: 53–65.
69 Funderburk SF, Marcellino BK, Yue Z: Cell
“self-eating” (autophagy) mechanism in Al-
zheimer’s disease. Mt Sinai J Med 2010;
77:
59–68.
70 Komatsu M, Waguri S, Chiba T, Murata S,
Iwata J, Tanida I, et al: Loss of autophagy in
the central nervous system causes neurode-
generation in mice. Nature 2006;
441: 880–
884.
71 Hara T, Nakamura K, Matsui M, Yamamoto
A, Nakahara Y, Suzuki-Migishima R, et al:
Suppression of basal autophagy in neural cells
causes neurodegenerative disease in mice.
Nature 2006;
441: 885–889.
72 Boland B, Kumar A, Lee S, Platt FM, Wegiel
J, Yu WH, et al: Autophagy induction and au-
tophagosome clearance in neurons: relation-
ship to autophagic pathology in Alzheimer’s
disease. J Neurosci 2008;
28: 6926–6937.
73 Rusten TE, Simonsen A: ESCRT functions in
autophagy and associated disease. Cell Cycle
2008;
7: 1166–1172.
74 Nixon RA: Autophagy, amyloidogenesis and
Alzheimer disease. J Cell Sci 2007;
120(pt 23):
4081–4091.
75 Helmfors L, Boman A, Civitelli L, Nath S,
Sandin L, Janefjord C, et al: Protective proper-
ties of lysozyme on β-amyloid pathology:
Implications for Alzheimer disease. Neuro-
biol Dis 2015;
83: 122–133.
76 Fuentes GP, Slachevsky CA: Enfermedad
de Alzheimer: actualización en terapia
farmacológica. Rev Med Chil 2005;
133:
224–230.
77 Roberson ED, Scearce-Levie K, Palop JJ, Yan
F, Cheng IH, Wu T, et al: Reducing endoge-
nous tau ameliorates amyloid beta-induced
deficits in an Alzheimer’s disease mouse
model. Science 2007;
316: 750–754.
78 Wang HY, Li W, Benedetti NJ, Lee DH: Alpha
7 nicotinic acetylcholine receptors mediate
beta-amyloid peptide-induced tau protein
phosphorylation. J Biol Chem 2003;
278:
31547–31553.
79 Zheng WH, Bastianetto S, Mennicken F, Ma
W, Kar S: Amyloid beta peptide induces tau
phosphorylation and loss of cholinergic neu-
rons in rat primary septal cultures. Neurosci-
ence 2002;
115: 201–211.
80 Götz J, Chen F, van Dorpe J, Nitsch RM: For-
mation of neurofibrillary tangles in P301l tau
transgenic mice induced by Abeta 42 fibrils.
Science 2001;
293: 1491–1495.
81 Stancu IC, Vasconcelos B, Terwel D, De-
wachter I: Models of β-amyloid induced Tau-
pathology: the long and “folded” road to un-
derstand the mechanism. Mol Neurodegener
2014;
9: 51.
82 Goedert M, Klug A, Crowther RA: Tau pro-
tein, the paired helical filament and Alzheim-
er’s disease. J Alzheimers Dis 2006;
9(3 suppl):
195–207.
83 Citron M: Alzheimer’s disease: strategies for
disease modification. Nat Rev Drug Discov
2010;
9: 387–398.
84 Kuret J, Congdon EE, Li G, Yin H, Yu X,
Zhong Q: Evaluating triggers and enhancers
of tau fibrillization. Microsc Res Tech 2005;
67: 141–155.
85 Rafii MS, Aisen PS: Recent developments in
Alzheimer’s disease therapeutics. BMC Med
2009;
7: 7.
86 Stoothoff WH, Johnson GV: Tau phosphory-
lation: physiological and pathological conse-
quences. Biochim Biophys Acta 2005;
1739:
280–297.
87 Lin MT, Beal MF: Mitochondrial dysfunction
and oxidative stress in neurodegenerative dis-
eases. Nature 2006;
443: 787–795.
88 Gandhi S, Abramov AY: Mechanism of oxi-
dative stress in neurodegeneration. Oxid Med
Cell Longev 2012;
2012: 428010.
89 Praticò D: Oxidative stress hypothesis in Al-
zheimer’s disease: a reappraisal. Trends Phar-
macol Sci 2008;
29: 609–615.
90 Makhaeva GF, Lushchekina S V, Boltneva
NP, Sokolov VB, Grigoriev VV, Serebryakova
OG, et al: Conjugates of γ-carbolines and phe-
nothiazine as new selective inhibitors of bu-
tyrylcholinesterase and blockers of NMDA
receptors for Alzheimer disease. Sci Rep 2015;
5: 13164.
91 Zhao Y, Zhao B: Oxidative stress and the
pathogenesis of Alzheimer’s disease. Oxid
Med Cell Longev 2013;
2013: 316523.
92 Grivennikova VG, Vinogradov AD: Genera-
tion of superoxide by the mitochondrial
Complex I. Biochim Biophys Acta 2006;
1757:
553–561.
Sanabria-Castro/Alvarado-Echeverría/
Monge-Bonilla
Ann Neurosci 2017;24:46–54
DOI: 10.1159/000464422
54
93 Hirai K, Aliev G, Nunomura A, Fujioka H,
Russell RL, Atwood CS, et al: Mitochondrial
abnormalities in Alzheimer’s disease. J Neu-
rosci 2001;
21: 3017–3023.
94 Zhu X, Perry G, Moreira PI, Aliev G,
CashAD, Hirai K, et al: Mitochondrial ab-
normalities and oxidative imbalance in Al-
zheimer disease. J Alzheimers Dis 2006;
9:
147–153.
95 Korol’ TY, Korol’ SV, Kostyuk EP, Kostyuk
PG: Disruption of calcium homeostasis in Al-
zheimer’s disease. Neurophysiology 2008;
40:
385–392.
96 Magi S, Castaldo P, Macrì ML, Maiolino M,
Matteucci A, Bastioli G, et al: Intracellular cal-
cium dysregulation: implications for Al-
zheimer’s disease. Biomed Res Int 2016;
2016:
6701324.
97 Fonseca AC, Cardoso SM, Pereira CF:
Calcium and redox homeostasis in Al-
zheimer’s disease: a focus on the endoplas-
mic reticulum. Ther Targets Neurol Dis
2014;
1: 1–13.
98 Stutzmann GE, Caccamo A, LaFerla FM,
Parker I: Dysregulated IP3 signaling in corti-
cal neurons of knock-in mice expressing an
Alzheimer’s-linked mutation in presenilin1
results in exaggerated Ca2+ signals and al-
tered membrane excitability. J Neurosci
2004;
24: 508–513.
99 Hagenston AM, Rudnick ND, Boone CE,
Yeckel MF: 2-Aminoethoxydiphenyl-borate
(2-APB) increases excitability in pyramidal
neurons. Cell Calcium 2009;
45: 310–317.
100 Small DH: Dysregulation of calcium homeo-
stasis in Alzheimer’s disease. Neurochem
Res 2009;
34: 1824–1829.
101 Wang JM, Sun C: Calcium and neurogenesis
in Alzheimer’s disease. Front Neurosci 2010;
4: 194.
102 Green KN, Smith IF, Laferla FM: Role of cal-
cium in the pathogenesis of Alzheimer’s dis-
ease and transgenic models. Subcell Bio-
chem 2007;
45: 507–521.
103 Arispe N, Rojas E, Pollard HB: Alzheimer
disease amyloid beta protein forms calcium
channels in bilayer membranes: blockade by
tromethamine and aluminum. Proc Natl
Acad Sci U S A 1993;
90: 567–571.
104 Crompton M, Barksby E, Johnson N,
Capano M: Mitochondrial intermem-
brane junctional complexes and their
involvement in cell death. Biochimie 2002;
84: 143–152.
105 Giorgio V, Soriano ME, Basso E, Bisetto E,
Lippe G, Forte MA, et al: Cyclophilin D in
mitochondrial pathophysiology. Biochim
Biophys Acta 2010;
1797: 1113–1118.
... Clinically, AD is characterized by abnormal aggregation of amyloid beta (Aβ) plaques extracellularly and hyperphosphorylated tau-derived intracellular neurofibrillary tangles in the hippocampus and cortex regions of the brain. Eventually, Aβ aggregation and tau phosphorylation lead to oxidative stress burden, synaptic downscaling, aggravated neuroinflammatory response, and, ultimately, neurodegeneration [1]. Before the onset of these pathological changes, an insulin-resistant brain state has been proposed to be a crucial factor in determining the disease progression. ...
... Likewise, another study suggested that MST1/Hippo signaling could be one of the therapeutic pathways for averting underlying neurodegeneration in AD [18]. The small molecule 4-((5,10-dimethyl-6-oxo-6,10-dihydro-5H-pyrimido [5,4-b]thieno [3,2-e] [1,4] diazepin-2-yl)amino)benzenesulfonamide or Xmu-mp-1 is recently discovered as a novel pharmacological inhibitor that antagonizes the activity of MST1/2 in a reversible and specific manner [19]. Xmu-mp-1 administration via intraperitoneal injection has been shown to have protective effects in sub-arachnoid and intracerebral hemorrhage, suggesting that Xmu-mp-1 manages to cross the blood-brain-barrier [20,21]. ...
... By providing the pioneering proof-of-concept for employing Xmu-mp-1 to alleviate AD pathology via modulation of the Hippo-Wnt signaling crosstalk, this study serves as a cornerstone for future investigations aimed at comprehending the pivotal interplay between the Hippo and Wnt signaling pathways in the light of AD therapeutics. ThioflavinT; Tnf-α: Tumour necrosis factor-α; Xmu-mp-1: 4-((5,10-Dimethyl-6-oxo-6,10-dihydro-5H-pyrimido [5,4-b]thieno [3,2-e] [1,4]diazepin-2-yl)amino)benzenesulfonamide; YAP: Yes-associated protein Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH ("Springer Nature"). Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users ("Users"), for smallscale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. ...
Article
Full-text available
Alzheimer’s disease (AD), the most prevalent form of dementia, is characterized by progressive cognitive impairment accompanied by aberrant neuronal apoptosis. Reports suggest that the pro-apoptotic mammalian set20-like kinase 1/2 (MST1/2) instigates neuronal apoptosis via activating the Hippo signaling pathway under various stress conditions, including AD. However, whether inhibiting MST1/2 has any therapeutic benefits in AD remains unknown. Thus, we tested the therapeutic effects of intervening MST1/2 activation via the pharmacological inhibitor Xmu-mp-1 in a sporadic AD rat model. Sporadic AD was established in adult rats by intracerebroventricular streptozotocin (ICV-STZ) injection (3 mg/kg body weight). Xmu-mp-1 (0.5 mg/kg/body weight) was administered once every 48 h for two weeks, and Donepezil (5 mg/kg body weight) was used as a reference standard drug. The therapeutic effects of Xmu-mp-1 on ICV-STZ rats were determined through various behavioral, biochemical, histopathological, and molecular tests. At the behavioral level, Xmu-mp-1 improved cognitive deficits in sporadic AD rats. Further, Xmu-mp-1 treatment reduced STZ-associated tau phosphorylation, amyloid-beta deposition, oxidative stress, neurotoxicity, neuroinflammation, synaptic dysfunction, neuronal apoptosis, and neurodegeneration. Mechanistically, Xmu-mp-1 exerted these neuroprotective actions by inactivating the Hippo signaling while potentiating the Wnt/β-Catenin signaling in the AD rats. Together, the results of the present study provide compelling support that Xmu-mp-1 negated the neuronal dysregulation in the rat model of sporadic AD. Therefore, inhibiting MST/Hippo signaling and modulating its crosstalk with the Wnt/β-Catenin pathway can be a promising alternative treatment strategy against AD pathology. This is the first study providing novel mechanistic insights into the therapeutic use of Xmu-mp-1 in sporadic AD.
... Symptoms of the late onset of AD often begin to manifest in the majority of individuals in their mid-60s. Early onset of Alzheimer's disease can also be seen between the age of 30 and 60, in rare cases (Sanabria-Castro et al. 2017). Memory loss, verbal difculty, and unpredictable behavior are the major symptoms associated with AD. ...
... According to the amyloid hypothesis, various enzymes such as -secretase (BACE1) and -secretase are responsible for the production of A from the cleavage of amyloid precursor protein (APP) (Sanabria-Castro et al. 2017). A reduction in the breakdown of A also leads to its accumulation in neurons and the amyloid plaques that cause neurodegeneration (Tabaton and Piccini 2005). ...
... Tau protein is a phosphoprotein that is connected to microtubules. In AD, tau protein has a pathogenic involvement mainly because of two reasonsposttranslational modications (Bakota and Brandt 2016) and abnormal phosphorylation of tau (Sanabria-Castro et al. 2017). Hyperphosphorylated tau forms due to aberrant phosphorylation of tau. ...
Chapter
Full-text available
The two major neurodegenerative diseases, viz., Parkinson’s disease (PD) and Alzheimer’s disease (AD), are incurable so far. These diseases are even difficult to manage as their exact pathogenesis is not clear, and currently the drugs that are being used for their management are just for symptomatic relief. At present, pathological pathways for these diseases are being explored at the molecular level, so that effective drugs can be discovered for the specific molecular targets. Recently, capsaicin (CAP) has been found to be effective in ameliorating some of the root causes of AD and PD, targeting some similar and some different mechanisms of action. In case of PD, dopaminergic neurons are lost and it has been found that CAP being a transient receptor potential vanilloid subtype 1 (TRPV1) agonist can help in the treatment of PD as it can further reduce oxidative stress and metabolic impairments of α-synuclein and inhibit glial activation and inflammation, protecting dopaminergic neurons in the striatum and substantia nigra. On the other hand, in the case of AD, amyloid-β (Aβ) accumulation is majorly reported to be responsible for the disease. It is suggested that inducing autophagy of Aβ can improve neural signaling in the brain. The CAP molecule has been explored for this action and has been found to attenuate Aβ amyloid, to improve locomotion, alleviating cognitive impairment. Research indicates that capsaicin, also has potential in combating one of the precursors to Alzheimer’s disease, ischemia. This suggests that it might contribute in reducing the symptoms associated with the condition. It was also found to decrease proinflammatory responses and oxidative stress in various AD models. Thus, it can be suggested that CAP may be utilized as a potential drug molecule against the two neurodegenerative disorders. The present chapter discusses the basic mechanisms behind these two diseases and further details on the role of capsaicin in their amelioration. In addition, certain limitations related to the therapeutic uses of CAP have been discussed.
... Alzheimer's disease (AD) is a neurodegenerative disease whose physiological and pathological mechanisms are not fully understood. 57,58 One important hallmark of AD is abnormal accumulation of beta-amyloid (Aβ) plaques and hyperphosphorylated tau tangles. [59][60][61] In 2022, Vagenknecht detected the abnormal accumulation of tau protein in AD mouse model by using volumetric multispectral photoacoustic tomography and a specific probe (i.e., PBB5), with a spatial resolution of 110 µm ( Figure 6B). ...
Article
Full-text available
Over the past few decades, the number of patients with neurological diseases has increased significantly, posing huge challenges and opportunities for the development of brain imaging technology. As a hybrid imaging method combining optical excitation and acoustic detection techniques, photoacoustic tomography (PAT), has experienced rapid development, due to high optical contrast and spatial resolution at depth inside tissues. With the development of lasers, ultrasonic detectors, and data computations, PAT has been widely applied for the diagnosis of oncology, dermatosis, etc. However, the energy of light and ultrasound would be greatly attenuated while penetrating the skull, due to the reflection, absorption, and scattering effects, resulting in limited application of PAT in brain imaging. In this review, we summarized the achievements of PAT and its application in the detection of brain diseases including glioma, stroke, traumatic brain injury, Alzheimer's disease, epilepsy, and Parkinson's disease. Moreover, various PAT systems and multi‐modality photoacoustic imaging are introduced for potential clinical applications. Finally, the challenges and current limitations of PAT for further brain imaging are also discussed.
... There are various well-established neurodegenerative mechanisms contributing to the onset and course of AD, such as β-amyloid plaque deposition, abnormal translocation of protein tau, deregulation of Rho and p-21-activated kinases, activation of inflammatory mediators, and a decrease in acetylcholine levels [11,12]. In addition to the several pathogenic processes that lead to the onset of AD, extensive abnormalities in insulin and insulin-like growth factor (IGF) signaling apparently play a critical role in the development of AD and the functioning of the brain [13]. ...
... In addition to various additional pathways including inflammatory reactions, oxidative stress, disturbance of calcium channels and autophagy are described to play a part in the progression of AD [59]. Mutations in APP, PSEN1, and PSEN2 are recognized inherited risk factors for AD [60][61]. According to Tanggis et al. (2012), FA can oxidatively hydroxy methylate pyrimidine bases in DNA, turn them into methyl groups and suppresses the expression of certain genes [62]. ...
... Metabolic disorders and reprogramming associated with aging may contribute to neurodegeneration and cancer development. Both ailments are related to pathways and genes concerned in bioenergetics, inflammation, DNA harm and repair, oxidative stress and unusual cell cycle activation [47,50]. There are several other factors that contribute to both conditions, such as obesity, diabetes, physical inactivity, smoking and family history. ...
Article
Background: Some preceding researches have observed that certain neurological disorders, such as Alzheimer's disease and multiple sclerosis, may affect breast cancer risk. However, whether there are causal relationships between these neurological conditions and breast cancer is inconclusive. This study was designed to explore whether neurological disorders affected the risks of breast cancer overall and of the two subtypes (ER+ and ER-). Methods: In the course of this study, genome-wide association study (GWAS) data for nine neurological diseases (Alzheimer's disease, multiple sclerosis, Parkinson's disease, myasthenia gravis, generalized epilepsy, intracerebral haemorrhage, cerebral atherosclerosis, brain glioblastoma, and benign meningeal tumour) were collected from the Complex Trait Genetics lab and the MRC Integrative Epidemiology Unit, and single-nucleotide polymorphisms (SNPs) extensively associated with these neurological ailments had been recognized as instrumental variables (IVs). GWAS data on breast cancer were collected from the Breast Cancer Association Consortium (BCAC). Two-sample Mendelian randomization (MR) analyses as well as multivariable MR analyses were performed to determine whether these SNPs contributed to breast cancer risk. Additionally, the accuracy of the results was evaluated using the false discovery rate (FDR) multiple correction method. Both heterogeneity and pleiotropy were evaluated by analyzing sensitivities. Results: According to the results of two-sample MR analyses, Alzheimer's disease significantly reduced the risks of overall (OR 0.925, 95% CI [0.871-0.982], P = 0.011) and ER+ (OR 0.912, 95% CI [0.853-0.975], P = 0.007) breast cancer, but there was a negative result in ER- breast cancer. However, after multiple FDR corrections, the effect of Alzheimer's disease on overall breast cancer was not statistically significant. In contrast, multiple sclerosis significantly increased ER+ breast cancer risk (OR 1.007, 95% CI [1.003-1.011], P = 0.001). In addition, the multivariable MR analyses showed that Alzheimer's disease significantly reduced the risk of ER+ breast cancer (IVW: OR 0.929, 95% CI [0.864-0.999], P=0.047; MR-Egger: OR 0.916, 95% CI [0.846-0.992], P=0.031); however, multiple sclerosis significantly increased the risk of ER+ breast cancer (IVW: OR 1.008, 95% CI [1.003-1.012], P=4.35×10-4; MR-Egger: OR 1.008, 95% CI [1.003-1.012], P=5.96×10-4). There were no significant associations between the remainder of the neurological diseases and breast cancer. Conclusions: This study found the trends towards a decreased risk of ER+ breast cancer in patients with Alzheimer's disease and an increased risk in patients with multiple sclerosis. However, due to the limitations of Mendelian randomization, we cannot determine whether there are definite causal relationships between neurological diseases and breast cancer risk. For conclusive evidences, more prospective randomized controlled trials will be needed in the future.
... Characterized by memory deficit, cognitive decline, and behavior change, AD is considered a progressive neurodegenerative disease [2]. Its pathology involves the formation of amyloid-beta (Aβ) and hyperphosphorylated tau protein (p-tau) [3,4]. While previous studies focused on targeting Aβ pathology for potential AD therapy, clinical evidence suggests limited effectiveness in slowing disease progression. ...
Article
Full-text available
Neurodegenerative disorders entail a progressive loss of neurons in cerebral and peripheral tissues, coupled with the aggregation of proteins exhibiting altered physicochemical properties. Crucial to these conditions is the gradual degradation of the central nervous system, manifesting as impairments in mobility, aberrant behaviors, and cognitive deficits. Mechanisms such as proteotoxic stress, neuroinflammation, oxidative stress, and programmed cell death contribute to the ongoing dysfunction and demise of neurons. Presently, neurodegenerative diseases lack definitive cures, and available therapies primarily offer palliative relief. The integration of nanotechnology into medical practices has significantly augmented both treatment efficacy and diagnostic capabilities. Nanoparticles, capable of traversing the blood–brain barrier, hold considerable potential for diagnosing and treating brain pathologies. By combining gene therapy with nanotechnology, the therapeutic effectiveness against neurodegenerative diseases can be substantially enhanced. Recent advancements in nano-biomaterial-based methodologies have fortified existing approaches to neural stem cell (NSC) differentiation therapies. NSC-targeting technologies offer a promising, potentially safe method for treating neurodegenerative diseases. This review endeavors to summarize current insights and perspectives on nanotechnology-driven therapeutic innovations in neurodegenerative disorders, with a particular emphasis on Alzheimer’s and Parkinson’s disease.
Article
Full-text available
Alzheimer’s Disease (AD) is a neurodegenerative disorder characterized by progressive neuronal loss. AD is associated with aberrant processing of the amyloid precursor protein, which leads to the deposition of amyloid- β plaques within the brain. Together with plaques deposition, the hyperphosphorylation of the microtubules associated protein tau and the formation of intraneuronal neurofibrillary tangles are a typical neuropathological feature in AD brains. Cellular dysfunctions involving specific subcellular compartments, such as mitochondria and endoplasmic reticulum (ER), are emerging as crucial players in the pathogenesis of AD, as well as increased oxidative stress and dysregulation of calcium homeostasis. Specifically, dysregulation of intracellular calcium homeostasis has been suggested as a common proximal cause of neural dysfunction in AD. Aberrant calcium signaling has been considered a phenomenon mainly related to the dysfunction of intracellular calcium stores, which can occur in both neuronal and nonneuronal cells. This review reports the most recent findings on cellular mechanisms involved in the pathogenesis of AD, with main focus on the control of calcium homeostasis at both cytosolic and mitochondrial level.
Article
Full-text available
Standardized extracts of Bacopa monniera (BME) have been shown to exert a neuroprotective effect against mental diseases, such as depression, anxiety and Alzheimer's disease (AD), in chronic administration studies. However, its mechanism of action has remained unclear. In this study, we evaluated the therapeutic effect of Bacopaside I (BS-I), a major triterpenoid saponin of BME, on the cognitive impairment and neuropathology in APP/PS1 transgenic mice and explored the possible mechanism from a biological systems perspective. We found that BS-I treatment significantly ameliorated learning deficits, improved long-term spatial memory, and reduced plaque load in APP/PS1 mice. We constructed BS-I's therapeutic effect network by mapping the nodes onto the protein-protein interaction (PPI) network constructed according to their functional categories based on genomic and proteomic data. Because many of the top enrichment categories related to the processes of the immune system and phagocytosis were detected, we proposed that BS-I promotes amyloid clearance via the induction of a suitable degree of innate immune stimulation and phagocytosis. Our research may help to clarify the neuroprotective effect of BME and indicated that natural saponins target the immune system, which may offer new research avenues to discover novel treatments for AD.
Article
Full-text available
The hallmarks of Alzheimer disease are amyloid-β plaques and neurofibrillary tangles accompanied by signs of neuroinflammation. Lysozyme is a major player in the innate immune system and has recently been shown to prevent the aggregation of amyloid-β1-40in vitro. In this study we found that patients with Alzheimer disease have increased lysozyme levels in the cerebrospinal fluid and lysozyme co-localized with amyloid-β in plaques. In Drosophila neuronal co-expression of lysozyme and amyloid-β1-42 reduced the formation of soluble and insoluble amyloid-β species, prolonged survival and improved the activity of amyloid-β1-42 transgenic flies. This suggests that lysozyme levels rise in Alzheimer disease as a compensatory response to amyloid-β increases and aggregation. In support of this, in vitro aggregation assays revealed that lysozyme associates with amyloid-β1-42 and alters its aggregation pathway to counteract the formation of toxic amyloid-β species. Overall, these studies establish a protective role for lysozyme against amyloid-β associated toxicities and identify increased lysozyme in patients with Alzheimer disease. Therefore, lysozyme has potential as a new biomarker as well as a therapeutic target for Alzheimer disease. Copyright © 2015. Published by Elsevier Inc.
Article
Full-text available
Alzheimer disease is a multifactorial pathology and the development of new multitarget neuroprotective drugs is promising and attractive. We synthesized a group of original compounds, which combine in one molecule γ-carboline fragment of dimebon and phenothiazine core of methylene blue (MB) linked by 1-oxo- and 2-hydroxypropylene spacers. Inhibitory activity of the conjugates toward acetylcholinesterase (AChE), butyrylcholinesterase (BChE) and structurally close to them carboxylesterase (CaE), as well their binding to NMDA-receptors were evaluated in vitro and in silico. These newly synthesized compounds showed significantly higher inhibitory activity toward BChE with IC50 values in submicromolar and micromolar range and exhibited selective inhibitory action against BChE over AChE and CaE. Kinetic studies for the 9 most active compounds indicated that majority of them were mixed-type BChE inhibitors. The main specific protein-ligand interaction is π-π stacking of phenothiazine ring with indole group of Trp82. These compounds emerge as promising safe multitarget ligands for the further development of a therapeutic approach against aging-related neurodegenerative disorders such as Alzheimer and/or other pathological conditions.
Article
Alzheimer's disease (AD) is a neurodegenerative condition characterized by increased accumulation of Aβ and degeneration of cholinergic signaling between basal forebrain and hippocampus. Nicotinic acetylcholine receptors (nAChRs) are important mediators of cholinergic signaling in modulation of learning and memory function. Accumulating lines of evidence indicate that a nAChR subtype, α7 receptor (α7-nAChR), plays an important role in modulations of excitatory neurotransmitter release, improvement of learning and memory ability, and enhancement of cognitive function. Importantly, the expression and function of α7-nAChRs is altered in the brain of AD animal models and AD patients, suggesting that this nAChR subtype participates in AD pathogenesis and may serve as a novel therapeutic target for AD treatment. However, the mechanisms underlying the role of α7-nAChRs in AD pathogenesis are very complex, and either neuroprotective effects or neurotoxic effects may occur through the α7-nAChRs. These effects depend on the levels of α7-nAChR expression and function, disease stages, or the use of α7-nAChR agonists, antagonists, or allosteric modulators. In this chapter, we summarize recent progresses in the roles of α7-nAChRs played in AD pathogenesis and therapy.
Article
The history of Alzheimer's disease (AD) presents a difficulty in a book that is devoted to the cholinergic system. Indeed, this history emphasizes the tale of plaques and bundles; the heroes of the tale are Alois Alzheimer and Gaetano Perusini, and their discoveries concern the amyloid cement; this tale, told in section 1 of this introduction, does not seem to relate to cholinergicity. Similarly, as a whole, the chapter focuses on neurocellular degeneration and degenerative proteins as the basis of AD rather than on cholinergicity. Yet, there is a strong link between AD and the cholinergic system in view of the apparent preference of plaques, bundles, and degenerative proteins for the central cholinergic neurons. This matter and its consequences are discussed in section 2 of this introduction and expanded upon in section K (Wong et al., 2006). © 2007 Springer Science+Business Media, LLC. All rights reserved.
Article
People who died of the neurodegenerative condition Creutzfeldt-Jakob disease after treatment with cadaver-derived human growth hormone also developed some of the pathological traits of Alzheimer's disease. See Letter p.247 Treatment of children with human cadaver-derived growth hormone (hGH) contaminated with prions resulted in iatrogenic transmission of Creutzfeldt–Jakob disease (iCJD). While such treatment ceased in 1985, CJD has a long incubation time and new cases are still emerging. Unexpectedly, an autopsy study of eight such iCJD patients, aged 36–51 years old, has revealed amyloid-β deposition in the grey matter typical of that seen in Alzheimer's disease and Aβ in the blood vessel walls characteristic of cerebral amyloid angiopathy. None of these patients had pathogenic mutations or high-risk alleles associated with early onset Alzheimer's disease. Importantly, Aβ pathology was detected only in prion-infected individuals who received hGH. This is consistent with iatrogenic transmission of Aβ pathology in addition to CJD, and suggests that healthy hGH exposed individuals may also be at risk of iatrogenic Alzheimer's disease and cerebral amyloid angiopathy.