ArticlePDF Available

Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis, New Perspectives for Pharmacological Treatment

Authors:

Abstract and Figures

Dementia is characterized by the impairment of cognition and behavior of people over 65 years. Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in the world, as approximately 47 million people are affected by this disease and the tendency is that this number will increased 62% by 2030. Two microscopic features assist in the characterization of the disease, the amyloid plaques and neurofibrillary agglomerates. All these factors are responsible for the slow and gradual deterioration of memory that affect language, personality or cognitive control. For the AD diagnosis, neuropsychological tests are performed in different spheres of cognitive functions but since not all cognitive functions may be affected, cerebrospinal fluid biomarkers are used along with these tests. To date, cholinesterase inhibitors are used as treatment, they are the only drugs that have shown significant improvements in the cognitive functions of AD patients. Despite the proven effectiveness of cholinesterase inhibitors, an AD carrier, even while being treated, is continually subjected to progressive degeneration of the neuronal tissue. For this reason, other biochemical pathways associated with the pathophysiology of AD have been explored as alternatives to the treatment of this condition such as inhibition of β-secretase and glycogen synthase kinase-3β. The present study aims to conduct a review of the epidemiology, pathophysiology, symptoms, diagnosis and treatment of Alzheimer's disease, emphasizing the research and development of new therapeutic approaches.
Content may be subject to copyright.
Send Orders for Reprints to reprints@benthamscience.ae
Current Medicinal Chemistry, 2017, 24, 1-19 1
REVIEW ARTICLE
0929-8673/17 $58.00+.00 © 2017 Bentham Science Publishers
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis,
New Perspectives for Pharmacological Treatment
Leide Caroline dos Santos Picançoa,*, Priscilla Farias Ozelaa, Maiara de Fátima de Brito Britoa,
Abraão Alves Pinheiroa, Elias Carvalho Padilhab, Francinaldo Sarges Bragac,
Carlos Henrique Tomich de Paula da Silvad, Cleydson Breno Rodrigues dos Santosc,
Joaquín María Campos Rosad,e and Lorane Izabel da Silva Hage-Melima,*
aLaboratório de Química Farmacêutica e Medicinal (PharMedChem), Universidade Federal do Amapá, Ma-
capá, Brazil; bFaculdade de Ciências Farmacêuticas, Universidade Estadual Paulista (UNESP), Campus
Araraquara, Departamento de Princípios Ativos Naturais e Toxicologia, Araraquara, São Paulo, Brazil;
cLaboratório de Modelagem e Química Computacional, Universidade Federal do Amapá, Macapá, Brazil;
dLaboratório Computacional de Química Farmacêutica, Departamento de Ciencias Farmaceuticas, Facul-
dade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo,
Brazil; eChemical Department of Pharmaceutical and Organic. Faculty of Pharmacy. University Campus of
cartuja. University Granada, 18071, Espanha
A R T I C L E H I S T O R Y
Received: August 13, 2016
Revised: November 20, 2016
Accepted: December 02, 2016
DOI: 10.2174/09298673236661612131
01126
Abstract: Dementia is characterized by the impairment of cognition and behavior of people
over 65 years. Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder in
the world, as approximately 47 million people are affected b y this disease and the tendency is
that this number will increase to 62% by 2030. Two microscopic features assist in the charac-
terization of the disease, the amyloid plaques and neurofibrillary agglomerates. All these fac-
tors are responsible for the slow and gradual deterioration of memory that affect language,
personality or cognitive control. For the AD diagnosis, neuropsychological tests are per-
formed in different spheres of cognitive functions but since not all cognitive functions may be
affected, cerebrospinal fluid biomarkers are used along with these tests. To date, cho linester-
ase inhibitors are used as treatment, they are the only drugs that have shown significant im-
provements in the cognitive functions of AD patients. Despite the proven effectiveness of
cholinesterase inhibitors, an AD carrier, even while being treated, is continually subjected to
progressive degeneration of the neuronal tissue. For this reason, other biochemical pathways
associated with the pathophysiology of AD have been explored as alternatives to the treatment
of this condition such as inhibition of β-secretase and glycogen synthase kinase-3β. The pre-
sent study aims to conduct a review of the epidemiology, pathophysiology, symptoms, diag-
nosis and treatment of Alzheimer's disease, emphasizing the research and development of new
therapeutic approaches.
Keywords: Alzheimer's disease, pathophysiology, treatment, acetylcholinesterase, β-secretase, glycogen synthase
kinase-3β.
1. INTRODUCTION
Dementia is characterized by impairment of cogni-
tion and behavior of people over 65 years of age. Alz-
*Address correspondence to this author at the Laboratório de Quí-
mica Farmacêutica e Medicinal, Universidade Federal do Amapá,
68902-280, Macapá, Brazil; Tel/Fax: +55 96 4009 2921;
E-mails: leidecaroline@outlook.com; lorane@unifap.br
heimer's disease (AD) is the neurodegenerative disor-
der most prevalent in the world, affecting about 24 mil-
lion people and it is estimated that by 2050 this number
will be quadrupled. Its main features are the deposit of
β-amyloid (Abeta) peptides in the extracellular surface
of neurons and the formation of neurofibrillary tangles
arising from the intracellular accumulation of hyper-
phosphorylated Tau protein.
2 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
AD is also associated with the deficit of the neuro-
transmitter acetylcholine (ACh) and oxidative stress
caused by exacerbation of glutamatergic transmission
[1-5]. In this review we will discuss the epidemiology,
pathophysiology, diagnosis, symptoms and prospects
for new pharmacological treatments for AD.
2. EPIDEMIOLOGY
Estimates in 2015 showed that around 46.8 million
people are affected by dementia worldwide. This num-
ber of new cases is almost 30% (9.9 million new cases)
greater than the incidence presented in the report of the
World Health Organization (WHO) in 2010. The high-
est incidence rates were in Asia (49%), Europe (25%)
and America (18%) [6, 7].
The prediction is that, by the year 2030, the number
will reach 74.7 million people and 131.5 million in
2050. Also, according to the estimates by the World
Alzheimer Report 2015 [6], East Asia and Africa are
the regions with the largest number of people with de-
mentia (about 9.8 million people), followed by Western
Europe with 7.4 million affected [6].
According to the Diagnostic and Statistical Manual
of Mental Disorders (2014) [8], the AD has a progres-
sive increase proportional to aging. Thus, the age is
considered to be a prevalence factor. Epidemiologi-
cally, AD affects approximately 5% of individuals over
65 years and 20% of those over 80 years. This means
that the rate of prevalence doubles every 5 years [9].
The World Alzheimer Report (2015) [6] pointed out
that in Europe and the Americas the highest incidence
of AD is between 80-89 years, in Asia among those
aged 75-84 years and 65-74 years in Africa.
Each year, in the United States, there is an increase
in the prevalence of AD in military veterans who have
suffered traumatic brain injury (TBI), post-traumatic
stress disorder and/or injuries associated with military
service [10]. These increasing infirm military veterans
tend to exacerbate hospital costs because of the need
for medical care and/or hospitalization for long periods.
In the wars of Iraq and Afghanistan, about 23% of
cases of TBI were originated from improvisation with
explosives [11].
People who have had moderate to severe TBI have a
two- to four-time higher risk to develop AD at older
ages [12]. A correlation with dementia development in
boxers was also observed [13]. But other sports of
much contact and strength, such as American football
and hockey, lead such athletes to TBI (about 1.6 to 3.8
million affected annually) in the United States [14-16].
The census of 2012 indicated that the population ob-
tained a substantial increase in the elderly population.
One indicator in this regard is the aging index, which is
the ratio of the number of people aged over 60 for
every 100 people under 15 years old. The rate in Brazil
is around 51.8, therefore the data show that there is a
person aged 60 years or older for every two people un-
der 15 years old [17]. In Brazil, more concrete data on
the AD come from the 2000s, when the statistical bases
of the country compared with data from other regions
revealed the prevalence in the country of 1.2 million
cases, and the incidence of 100,000 new cases each
year [18]. However, there is need to improve the search
and registration of the data, since the AD may be un-
registered or underdiagnosed [19, 20].
According to the World Alzheimer Report 2015 [6],
the overall costs of the disease (medical care, social
care and informal care) were 818 billion dollars with an
increase of 35.4% compared to the same study in 2010
[7]. The trend of the cost with Alzheimer's disease for
2018 will be $ 1 trillion, rising to 2 trillion in 2030 [6].
3. PATHOPHYSIOLOGY
Even with over a hundred years of history, the AD
does not have full clarification regarding its pathogene-
sis and still lacks a therapy that induces a natural heal-
ing. Nevertheless, macroscopic and microscopic mark-
ers related to it are known and may help in its charac-
terization, in understanding of the disease pathogenesis
and in the development of possible strategies [21, 22].
At the macroscopic level (Fig. 1) there is the atro-
phy of the hippocampus and cerebral cortex, which in
AD appears more sharply due to age [23, 24]. Micro-
scopically it is possible to observe the formation of
amyloid plaques, or senile plaques, which are amor-
phous structures of Abeta, and accumulation of hyper-
phosphorylated Tau protein which implies the forma-
tion of neurofibrillary tangles, and extensive neuronal
loss [25-30].
Recent research showed other mechanisms are re-
lated to the formation of these AD markers, ranging
from genetic imprint factors [30, 32] as family heritage
[33], and mechanisms that involve apolipoprotein E
[27, 31, 34], the mechanism of oxidation processes [25,
35] culminating in the neurodegeneration process.
3.1. Genetic Mechanism
The vast majority of AD case occurrences are spo-
radic, i.e. where there is no dominant genetic cause, but
rare mutations may occur in the APP gene, originating
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 3
the familiar AD [33], and allele ε4 of apolipoprotein E
(ApoE) that has been shown in recent studies as the
strongest genetic risk factor related to AD development
process [27, 31].
Fig. (1). Macroscopic changes. Atrophy of the hippocampus
and cerebral cortex.
The human carriers of ApoE are present in ap-
proximately 1 of every 5 individuals. The subsidy that
ApoE provides to the development of AD is realized
when it is observed that these individuals account for
about 65% of cases of the disease [31] and that carriers
of the ApoE have the risk of developing Alzheimer's
increased three fold [32]. The mechanisms that corre-
late the presence of ApoE with AD are not well under-
stood yet, however, it is being suggested that in such
cases there is a decrease in clearance of Abeta in the
brain [31, 36].
In addition to the mechanism involving ApoE, stud-
ies also demonstrate a strong correlation between the
presence of preselinin alleles (PSEN 1 and PSEN 2) in
AD-patients and in individuals with a tendency to be
carriers of AD or other related diseases [37, 38]. There
is precedence in literature of the identification of two
different types of mutation in this gene in patients with
familial AD [39, 40].
PSEN 1 and PSEN 2 genes are rare in individuals
with AD, although their relationship with ApoE is a
major factor when the disease is due to genetic causes.
They are involved in early-onset pathology, which is a
rare form of the disease [41]. Mutations in PSEN 1
(chromosome 14) account for 18% to 50% of these
early onset cases [42], mutations in PSEN 2 (chromo-
some 1) have been rarely reported and was mostly de-
tected in African populations and in European coun-
tries [43]. The mechanism of correlation between pre-
selinin and the control of APP cleavage has not yet
been fully elucidated [44].
3.2. Amyloid hypothesis and Protein Tau
As previously mentioned, the Abeta fragments and
neurofibrillary tangles are important markers for AD
[25-27] which characterize the amyloid hypothesis.
These deposits are the result of the wrong folding of
native proteins, i.e. forming after altered cleavage of
the amyloid precursor protein (APP) [45-47].
APP is a transmembrane glycoprotein with ap-
proximately 770 amino acids expressed by several
cells, including CNS neurons [47, 48]. The cleavage of
APP occurs through the enzymes α-, β- and ɣ-secretase
[44, 49], while the amyloid pathway has its cascade
unleashed when APP is cleaved by β-secretase thereby
forming insoluble peptides having 39 to 43 fragments
[47, 48, 50].
The Abeta fragments, especially the Abeta-42 iso-
form, have pronounced cytotoxic properties which are
related to the process of neurodegeneration, for facili-
tating the formation of oxiradicals [51], being or not
directly toxic to neuronal cells, by deregulating calcium
homeostasis due to lipidic dysregulation of the cell
membrane [26, 27, 36, 37, 52], these fragments form
the insoluble structures that characterize histopa-
thologically the AD (senile plaques) and this process
ends up leading to neuronal death [53, 54].
Tau protein promotes a kind of assembly of tubulin,
providing microtubule stability [33]. Neurofibrillary
tangles are the result of Tau protein hyperphosphoryla-
tion. According to the results presented in the work of
Stancu and co-workers, although mice models that ex-
press the mutant amyloid precursor protein without tau
overexpression did not show neurofibrillary tangles,
subtle changes in mice endogenous Tau were induced
by high concentrations of Abeta, resulting in the proc-
ess of hyperphosphorylation of Tau [27]. Research has
also found strong evidence of the interdependence be-
tween Aβ accumulation and tau protein aggregation,
which represents the final stage of disease pathogenesis
[55, 56].
3.3. Inflammatory Mechanism and Mitochondrial
Dysfunction
AD is a disease that is closely linked to inflamma-
tory processes [41, 57]. In addition, several studies
have shown that Tau pathologies are drastically exac-
erbated during the occurrence of acute and chronic in-
flammatory processes [27, 58-60]. Those inflammatory
processes are mediated or induced by microglial clus-
ters around the densest regions of Abeta plaques, by
high levels of pro-inflammatory cytokines and by mi-
4 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
croglial activation that precedes the formation of neu-
rofibrillary tangles (Fig. 2) [41].
With regard to mitochondrial dysfunction, it is be-
lieved that the deposit of Abeta fragments and patho-
logical Tau protein affect mitochondrial function in
brain cells, specifically in regard to the impairment of
mitochondrial oxidative metabolism [27, 35, 61]. Stud-
ies related to the presence of Abeta peptides carried out
by Benevento enabled him to conclude, with more
specificity, that these peptides can be directly toxic to
neuronal mitochondria [61].
3.4. Oxidative Stress
Studies show that there is strong evidence that oxi-
dative stress induced by Abeta is crucial to the patho-
genesis and progress of AD [26], being present, both as
cause and consequence, of inflammatory processes in
general, which is characteristic of neurodegenerative
diseases [62].
The brain is an organ that has high energetic activ-
ity, this energy demand is supplied by mitochondrial
oxidative phosphorylation, and this process can lead to
the formation of highly reactive oxygen species [63]:
oxidative stress is a result of excessive production of
these species. In this case, the protective mechanisms
are compromised, the reactive oxygen species begin to
accumulate and the neurons become susceptible to the
excitotoxic lesion [47]. However, this mechanism de-
pends on Abeta fragments which, when accumulated,
promote the reduction of iron and brain copper, which
are key factors to trigger oxidative stress [64] which,
under these conditions, promotes DNA damage [25].
3.5. Cholinergic Hypothesis
Among the mechanisms related to the onset and
evolution of AD, a extremely studied hypothesis is the
cholinergic hypothesis, which was the first theory re-
lated to AD pathogenesis [54, 64].
In general, the brain of AD carriers presents, in ad-
dition to the histopathological markers previously de-
scribed, atrophy, synaptic loss and deficiency in central
neurotransmission. There is overall degeneration of
basal forebrain neurons [65]. In the beginning of the
disease there is loss of the cholinergic neurons in the
basal nucleus and in the entorhinal cortex, but in the
advanced stage of AD, more than 90% of the choliner-
gic neurons of the basal nucleus are lost [66].
Bartus and Emerich (1999) [67] point out that ac-
cording to the cholinergic hypothesis, the abnormal or
impaired functioning of the cholinergic system proves
Fig. (2). Amyloid cascade in AD.
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 5
capable of inducing a memory deficiency in animal
models, similar to AD. According to this hypothesis,
the destruction of cholinergic neurons in the basal fore-
brain and the loss of central cholinergic transmission
leads to the appearance of cognitive and non-cognitive
symptoms in patients with AD [66].
It is also observed that the cholinergic hypothesis is
also characterized by: a marked decrease in acetyltrans-
ferase concentration responsible for the synthesis of
acetylcholine in the cortex and hippocampus; and due
to the loss of cholinergic neurons in the Meynert basal
nucleus [68]. The cholinergic hypothesis also corrobo-
rates that the dysfunction and cell death of neurons re-
sponsible for the maintenance of specific transmission
systems leads to the deficiency of Ach, noradrenaline
and serotonin [69].
4. SYMPTOMATOLOGY
AD is often a major cause of dementia in elderly
people, when the evolution of the symptoms becomes
severe, with slow and gradual deterioration of the
memory that affects language, personality and cogni-
tive control [70-74]. AD has major medical, economic
and social consequences due to problems such as de-
mentia, dependency and disability [75].
AD can be classified in three stages: the early
stage, where the loss of memory is discrete; cholinergic
neurons in the limbic system are affected, the hippo-
campus shrinks by approximately 25% of its volume.
The light declines in memory capacity are associated
with damaged neurons since they are responsible for
the short and long term memory; the intermediate
stage, which is characterized by difficulty in recogniz-
ing and communicating with people, is a long stage and
can last from two to ten years according to Seidl [76].
After that, there is a decline in the acetylcholine levels
of some neurons, among these are those located in the
ventral telencephalon, which under normal conditions
are involved in information storage and long-term
memory; and the final stage, in which the patient is
completely debilitated and there is total dependency
and inability to perform daily activities. Additionally,
no information recovery is possible due to the interfer-
ence of the disease in the limbic system. The destruc-
tion of stored memories are associated with degenera-
tion of cholinergic neurons throughout the cerebral cor-
tex. The person forgets his past, friends and family.
This phase lasts from one to three years on average and
ends with the death of the patient [77-82].
According to Petronilho, this condition is mainly as-
sociated to the reduction of acetylcholine (ACh) levels
in the synaptic process, since this neurotransmitter is
directly related to cognitive, motor and memory proc-
esses [83]. In the early stages of AD there are typical
behavioral changes such as irritability and indifference
in family affairs or entertainment [71]. Patients show
personality changes, confusion, anger, sadness, lack of
direction and difficulty in concentrating. Agitation is
associated with loss of volume in several specific areas
of the brain, including the frontal cortex, anterior cin-
gulate cortex (ACC), posterior cingulate cortex (PCC),
insula, amygdala and hippocampus. There are two dif-
ferent mechanisms that explain the agitation in AD,
one is due to emotional regulation deficits (emotional
responses) and another is due to executive function
deficits (problem solving). However, it is important to
state that their evolution may vary from patient to pa-
tient [84].
Over time, more than 90% of all patients feel
symptoms such as apathy, restlessness, anxiety, depres-
sion, hallucinations, delirium, motor activity abnormal-
ity, irritability, sleep disorders, eating disorders, eupho-
ria or disinhibition. Mood disorders affect a significant
percentage of individuals who develop AD at some
point in the evolution of dementia. Depressive symp-
toms are observed within 40-50% of patients, while
depressive disorders affect about 10- 20% of cases.
Depressive symptoms are very common in AD, major
depressive disorder has been extensively studied as a
possible neurocircuitry disorder, but there are still very
few neuroimaging data published involving depression
in AD [84-88].
AD patients develop a gradual and insidious cogni-
tive deficit that becomes disabling in advanced stages
of the disease. These devastating symptoms compro-
mise significantly the quality of life of patients, leading
to absolute dependence, hospitalization and inevitably
death [77, 89-91].
5. DIAGNOSIS
The diagnosis of AD performed in an initial stage of
the disease is essential to ensure that the patient has
good living conditions [92]. For this, the criteria of the
Diagnostic and Statistical Manual of Mental Disorders
(DSM IV) and established standards by the National
Institute of Neurological and Communicative Disorders
and Stroke (NINCDS) concomitant with Alzheimer's
Disease and Related Disorders Association (ADRDA)
are used [93]. These neuropsychological tests and psy-
chometric tests, in various fields, analyze the cognitive
functions of patients. In addition, blood tests, structural
neuroimaging, molecular and functional neuroimaging,
6 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
cerebrospinal fluid, electroencephalogram (EEG) and
evoked potentials are performed. Genetic studies are
also important complementary tests that can integrate a
more complete diagnosis of the disease [94].
For global cognitive function, the main clinical ex-
amination established by NINCDS-ADRDA is the
Mini-Mental State Examination (MMSE). The MMSE
is one of the most used tests in the world. It was pre-
pared by Folstein et al., [94]. This test can be used
alone or coupled with other tools to assess the cogni-
tive level of the patient [95]. Nitrini and colleagues
[96] recommend the association of Information -
Memory - Concentration Test (IMC) of Blessed [97]
and The Cognitive Abilities Screening Instrument -
Shortform [98] for ratification of the reduction in
global cognitive framework.
In the memory assessment, delayed recall tests can
be used [99], the Rey Auditory Verbal Learning Test
(RAVLT) [100] or the memory logic of the Wechsler
Memory Scale (WMS) [101]. In this aspect, exams in
Brazil also use a battery of neuropsychological delayed
recall of the Consortium to Establish a Registry for
Alzheimer's Disease (CERAD) [102].
Analyzing the cognitive function of attention, the
most applied exams are random letter Test [103], Digit
Extension (direct and reverse) and the Trail Making
Test [104].
To check the cognitive framework of language the
instrument ADAS-cog is used, naming real objects
[105]; Boston Naming Test (BNT) that applies the
name of 15 figures in black and white [106] or desig-
nating eight figures in the Abbreviated Neuropsy-
chological battery test (NEUROPSI) [107].
Regarding conceptualization and abstraction, the
Cambridge Examination for Mental Disorders Similari-
ties Test of the Elderly (CAMDEX) [108] or NEU-
ROPSI [109] are tests that require the ability to relate
three pairs of nouns.
In constructive skills, the patient is examined by the
Clock Drawing Test (CDT) [110] and geometric de-
signs of CERAD [102].
However, these neuropsychological tests have be-
come deadlocked by the new proposals of clinical
manifestations, such as mild cognitive impairment
(MCI) [111]. Loss of timely memory and cognitive
changes in routine cases can be common and not nec-
essarily an early development of AD, as many activi-
ties are still preserved [111, 112]. From this context, it
is necessary that additional tests reinforce the diagnosis
of AD [113].
Laboratory blood tests for diagnosis of AD should
include the following parameters: complete blood
count, serum urea, creatinine, thyroxine (T4), thyroid
stimulating hormone (TSH), albumin, liver enzymes
(SGOT, SGPT, gamma GT), vitamin B12, calcium,
serologic tests for syphilis and in patients younger than
60 years, complete HIV serology [114].
Computed tomography (CT) and magnetic reso-
nance imaging (MRI) are important structural neuroi-
maging exams. CT reveals subdural hematomas, tu-
mors or normal pressure hydrocephalus. However, de-
pending on the clinical history of the patient, these may
be causes of reversible dementia [94]. Therefore, brain
MRI details more accurately change in areas of the
cerebral cortex and hippocampus [106].
Cerebrospinal fluid exams analyze the changes in
two biomarkers: reduced Abeta protein 1-42 (Abeta42)
which is a component of neuritic plaques and increased
levels of Tau protein (total and phosphorylated) that is
related to neuronal decay [115-117].
The positron emission tomography (PET) identifies
markers of β-amyloid plaques (pathological signature)
deposited in neuronal tissue [118]. The tomography
single photon emission computed tomography
(SPECT) is used for differential diagnosis of dementia,
including depression of the elderly and differential di-
agnosis of Alzheimer's disease and dementia through
Lewi bodies (generalized in the cortex) [119].
In addition, there are molecular diagnostics associ-
ated with magnetic resonance spectroscopy (MRS).
MicroRNAs (miRNA), small non-coding RNAs that
regulate gene expression at post-transcriptional level,
have been applied to the diagnosis of AD [120].
Changes in miRNA network caused by the disease,
contribute to deregulation of genes of the amyloi-
dogenic cascade (genes such as APP, BACE1 and
MAPT) [121].
6. PHARMACOLOGICAL TREATMENT
6.1. Acetylcholinesterase Inhibitors
To date, cholinesterase inhibitors (Table 1) are the
only drugs that have shown significant improvements
in the cognitive process of patients with AD, to reduce
their symptoms by improving the cholinergic function
in neuronal synapses [122,123]. These drugs act as in-
hibitors of cholinesterase (Acetylcholinesterase -
AChE, and butyrylcholinesterase - BChE), which are
enzymes responsible for degradation of the neuro-
transmitter acetylcholine (ACh) in the synapses, after
transmission of the nervous impulse. Thus, to inhibit
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 7
them, cholinesterase inhibitors increase the availability
of these neurotransmitters in the synaptic cleft, reduc-
ing the symptoms of AD [123].
In this context, many studies have been conducted
in order to discover new drugs that act in this patho-
genic pathway. However, to date, only 4 drugs were
actually approved by regulatory agencies for the treat-
ment of AD, which are: Tacrine (Fig. 3a), Galantamine
(Fig. 3b), Rivastigmine (Fig. 3c) and Donepezil (Fig.
3d) [122].
N
NH2
O
O
O
N
O
O
N
HO
N
O
O
N
(a) (b)
(c) (d)
(e)
H2N
Fig. (3). FDA-approved drugs for the treatment of (a) tacrine, (b)
galantamine, (c) rivastigmine, (d) donepezil, (e) memantine [133,
134].
Tacrine was the first drug approved for the treat-
ment of AD. It is considered a non-competitive and
non-selective reversible inhibitor to acetylcho-
linesterase (AChE), which has a dose-dependent effi-
cacy, short half-life and high incidence of adverse ef-
fects and hepatotoxicity [124, 125].
Donepezil is a drug from the family of N-
benzylpiperidine; it was developed, synthesized and
evaluated by a Japanese pharmaceutical industry, and
was approved in 1996 for its use in the treatment
against AD. It is a highly selective reversible noncom-
petitive inhibitor of AChE, does not present serious
adverse effects and significantly improves the symp-
toms of AD [126, 128]. However, by using the cyto-
chrome P450 system in its metabolism, emerges the
development of drug interactions with other drugs, so
that their combination should be cautious [124].
Rivastigmine, a physostigmine derived-drug, is a
pseudo-selective irreversible inhibitor of AChE and
BChE. This drug shows good activity and tolerance in
patients with AD and does not involve the cytochrome
P450 system in its metabolism, reducing the possibility
of occurrence of drug-drug interactions, improving
cognition and generating neuroprotective effects [124,
129, 130].
Galantamine is a tertiary alkaloid extracted from
various species of Amaryllidaceae, accidentally dis-
covered in 1950. It is a selective, competitive and re-
versible inhibitor of AChE and contains a modulating
action on nicotinic receptors and low hepatotoxicity.
However, its association with other drugs requires cau-
tion since its metabolism uses the cytochrome P450
system. This drug was only approved in 2001 for the
treatment of AD, but it was used over the years for
various neurological disorders [124, 125, 131].
6.2. NMDA Antagonists
Memantine (l-amino-3,5-dimethyl adamantane)
(Fig. 3e) (Table 1) was approved in May 2002 by the
European Union and, in October 2003, was licensed by
the FDA (Food and Drug Administration). This drug
decreases excitotoxicity and neurodegeneration caused
by excessive action of glutamate [134, 135].
Memantine is a noncompetitive NMDA receptor an-
tagonist used for the treatment of AD in mild to severe
stages and presents a half-life less than 60 hours. Me-
mantine reduces excessive glutamatergic neurotrans-
mission, may decrease hyperphosphorylation of Tau
protein and protects against toxicity induced the Abeta
peptide [136, 139]. The blocking property of this recep-
tor was discovered by Kornhuber et al., [140] when
used as a selective inhibitor of the NMDA receptor, the
MK-801 compound.
In the 90s, the blocking action of memantine was
elucidated by Parsons et al., [141]. Memantine in vivo,
inhibits mitochondrial function, reduces both the cere-
bral blood flow and the toxic effects of the neuro-
inflammation, and the formation of Abeta peptide
[142]. Meta-analysis studies corroborate that the drug
reduces the neuropsychiatric symptoms of AD when
compared with a placebo [143]. However, Fox et al.,
[138] proved that the use of memantine does not sig-
nificantly improve the symptoms of agitation in people
with moderate to severe AD.
Hu et al., [144] found that in primary cultures of
granular neurons in the rat cerebellum, a derivative of
tacrine, such as bis(propyl)cognitin (B3C) (IC50 = 0.45
mM) proved to be 10 times more potent than mem-
antine (IC50 = 4.58 uM) to decrease the glutamate-
induced excitotoxicity. The B3C is a noncompetitive
8 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
Table 1. Anti-Alzheimer agents and mechanism of action.
Agents
Mechanism
References
1
Tacrine
Non-competitive and non-selective reversible inhibitor
to acetylcholinesterase
[125, 126]
2
Donepezil
Highly selective reversible noncompetitive inhibitor of
AChE
[127-129]
3
Rivastigmine
Pseudo-selective irreversible inhibitor of AChE and
BChE
[125, 130, 131]
4
Galantamine
Selective, competitive and reversible inhibitor of
AChE
[125, 126, 132]
5
Memantine
[137-140]
6
Bis(propyl)cognitin (B3C)
Non-competitive NMDA receptor antagonist
[146]
7
LY2811376
Block the enzymes that cleave the amyloid precursor
protein (APP)
Non-peptide BACE1 inhibitor
8
2,2',4'-trihydroxichalcone (TDC)
9
Ginsenoside Rg1
BACE1 inhibitor
[156]
10
OM99-2
Potent peptide inhibitor of leucine and alanine
(BACE1 inhibitor)
[157]
11
hydroxyethylamine isosteres (HEA)
12
Isophthalamide
[158]
13
des(dimethylamino)
BACE1 inhibitor
[159]
14
LY45013A
γ-secretase inhibitor
[153]
15
Thiazolidinones (TZD)
ATP-non-competitive inhibito r
16
Bis-indole
[171]
17
Aniline
[173]
18
Maleimides, Kenpaullone
[174]
19
Indirubin
[175]
20
Hymenialdisin
GSK-3β inhibitor
[176]
21
SB216763 (3-[2,4-dichlorophenyl]-4-
[1-methyl-1H-indol-3-yl] -1H-pyrrole-
2,5-dione)
Highly selective inhibitor that possesses cell perme-
ability and acts by competing with ATP binding site
[162, 168]
22
SB415286
Blocks the glutamate induced apoptosis
[145]
23
Lithium
Deprive potassium or inhibits the enzyme competing
with magnesium ions (Mg2+)
[179-181]
24
Valproate sodium
Reduced the phosphorylation of the Tau protein
[184]
25
Mts-L803
Inhibits Abeta peptide accumulation and improve the
cognitive function
[185]
26
Piperazinyl sulfonamide analogs
GSK-3β inhibitor
[186]
27
derivatives of 6-amino-4-(pyrimidin-
4-yl)pyridone
[187]
28
2-(2-phenylmorpholin-4-yl)pyrimidin-
4(3H)-ones
Decreased phosphorylation of the Tau protein
[170]
29
1,3,4-oxadiazole derivatives
Highly selective inhibitors of GSK-3β
[188-191]
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 9
(Table 1) contd….
Agents
Mechanism
References
30
Quinolone derivatives
Inhibitory activity against GSK-3β and neuroprotec-
tive action against the injury caused by the production
of Abeta peptide
[192]
31
Compounds C-7a and C-7b
Interfere with the neuronal death-induced accumula-
tion of Ab eta peptides and inhibits the phosphorylation
of the Tau protein
[194]
32
Pyrimidones
[195]
33
Np-1003
34
Methylthioninium chloride (TRx0237)
GSK-3β inhibitor
[196]
35
Indomethacin
36
Ibuprofen
Reduces formation of Abeta
[200]
37
Curcumine
Modulate eicosanoid biosynthesis and inhibits COX-1,
COX-2 and LOX
38
Magnolol
39
α-pinene
40
β-pinene
41
Carvacrol
42
Luteolin
43
Rosmarinic acid
44
Resveratrol
45
Apocynin
Inhibits COX
46
4’-O-metylhonokiol (MH)
Inhibits the induction of amyloidogenic
Inhibits the activation of astrocytes
[134, 197, 201, 202]
47
Quercetin
48
Myricetin
Antioxidant
[75]
49
AT-1792
Act on amyloid plaques
[198]
NMDA receptor antagonist that interacts only in patho-
logical situations as well as an AChE inhibitor and an-
tagonist of the γ-aminobutyric acid receptor subtype A
[145].
A rat model had their learning and memory func-
tions impaired by the okadaic acid, in this model the
impairment was reduced followed the administration of
memantine (10 mg/kg). It was assessed by the elapsed
time for the animal to find a submerged platform in the
Morris water maze (latency); it was significantly de-
creased after first session [146].
According Winblad et al., [147], memantine sho-
wed benefits for daily activities of patients with moder-
ate to severe DA when compared to a placebo, and thus
lead them to a dignified life. Patient groups (already
treated with acetylcholinesterase inhibitors) that re-
ceived only memantine or associated with tocopherol
showed no improvement in mild to moderate AD ac-
cording to Dysken et al., [148].
After 2 hours of memantine administration (50
mg/kg), there was increased phosphorylation at Ser9
residue from glycogen synthase kinase-3β (GSK-3β) in
the cerebral cortex of mice, thus inhibiting the enzyme
activity [137, 149]. In a study by Hellweg et al., [150]
in 2,506 patients with moderate to severe AD, indi-
viduals treated with memantine showed a significant
delay in worsening of clinical condition compared with
those who received placebo.
In the latency of the two-way avoidance response,
the mice treated with the drug memantine and the com-
10 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
pound FTY720 had a longer latency to enter the dark
compartment and showed less time spent inside the
compartment that the untreated animals. Histological
analysis of hippocampal neurons, showed that the ani-
mals that received FTY720 and memantine had a sig-
nificant reduction in neuronal loss induced by Abeta
peptide [151].
In the study of 882 patients, who received a placebo
and 868 patients were treated with memantine an im-
provement of 1.99 in the Neuropsychiatric Inventory
scale was observed when compared with the placebo
group [143].
6.3. Secretases Inhibitors
The secretase inhibitors (Table 1) block the en-
zymes that cleave the amyloid precursor protein (APP),
thereby preventing the formation of insoluble Abeta
peptides that are responsible for the formation of senile
plaques. The main enzymes participating in this patho-
genic mechanism are the β-secretase (BACE1) and γ-
secretase [152].
According to Sinha and Liebenburg [153], the β-
secretase is an interesting therapeutic target for AD,
presenting an essential role in the production of Abeta.
Thus, Zhu et al., [154] report that subsequent to the
BACE1 identification, several researchers have sought
synthesizing and testing numerous inhibitors of this
enzyme with regard to their Abeta reduction properties,
in an attempt to retard the progression of AD in the
long term.
Zhang [155] reported the LY2811376 as the first
non-peptide BACE1 inhibitor drug available for oral
administration in clinical trials. However, the studies
had to cease when a significant increase in toxicity of
the retinal epithelial cells of the animals studied was
detected, thus suggesting the need for greater care dur-
ing the planning, use and safety assessment of this type
of drugs.
Among the compounds used for inhibition of
BACE1, there are some naturally occurring compounds
used in traditional Chinese medicine, such as 2,2',4'-
trihydroxichalcone (TDC) and ginsenoside Rg1 [155],
and other synthetic compounds such as OM99-2 [156],
hydroxyethylamine isosteres (HEA), the isophthala-
mide [157] and a des(dimethylamino) compound [158].
Regarding the natural origin BACE1 inhibitors, the
Rg1 ginsenoside is obtained from Panax notoinseng. It
has been used extensively over the years in traditional
medicine for improving the memory function and pre-
sented about 80% inhibitory activity against BACE1,
revealing its potential protection against the degenera-
tive effects of Abeta. Since the DCT is a family of fla-
vonoid chalcones extracted from the Glycyrrhiza
glabra, widely used as an emollient in stomach disor-
ders and respiratory problems. It presents anti-
inflammatory, antioxidant and anti-tumor activities;
while more recently it has been evaluated as a potential
candidate for the treatment of AD due to a surprising
inhibitory activity against BACE1, suppressing the
cleavage of the amyloid precursor protein and thereby
reducing the levels of toxic Abeta peptide in brain tis-
sue [155].
The OM99-2, a hydroxyethylene-containing potent
peptide inhibitor of leucine and alanine, was the first
compound to act effectively on the BACE1 [156, 159],
while other studies of Gosh et al., [157] found that in
terms of structure-activity relationship (SAR), the sizes
of structures and the choice of substituents in the hy-
droxyethylamine moiety was crucial for the inhibitory
potency of the drug against BACE1, just as some bioi-
sosteres of isophthalamide had better inhibitory poten-
tial, with good cell inhibitory activity and considerable
reduction in the production of toxic Abeta peptides,
such as the GRL-7234 and GRL-8234.
In 2009, Steele et al., [158] have identified the
aminopyrimidine-containing molecule (HTS) as a weak
inhibitor of BACE1 (IC50 = 317 µM), and only after
several optimization experiments, by molecular and
rational design modeling, the HTS originated the
des(dimethylamino) chemical structure, which in terms
of SAR showed an enzymatic activity superior to HTS.
In addition to the drugs mentioned above, Simers
and colleagues [160] obtained good results in clinical
trials of LY45013A, an inhibitor of γ-secretase, which
during the phase I studies showed a dose-dependent
reduction in plasma levels of Abeta without important
adverse effects [152].
6.4. Glycogen Synthase Kinase Inhibitors:
The Glycogen Synthase Kinase-3 is a ser-
ine/threonine kinase protein responsible for the phos-
phorylation and thus inactivation of glycogen synthase,
the control of glycogen metabolism, regulation of cell
proliferation and cell cycle. There are two isoforms of
GSK-3, GSK-3α and GSK-3β. The first presents the α-
aminoacid glycine in its N-terminal end, whereas the
second consists of two splice variants, in which the
shorter form (GSK-3β 1) is in several organs and the
longer form (GSK-3β2) is present in the central nerv-
ous system [161-164].
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 11
GKS-3β activity is negatively regulated by phos-
phorylation of serine 9 (Ser9) and positively by phos-
phorylation of tyrosine 216 (Tyr216), i.e., the phos-
phorylation of Ser9 residue reduces the activity of this
enzyme, whereas the phosphorylation of Tyr216 resi-
due promotes activation [165-167].
The activation of GSK-3β causes the hyperphos-
phorylation of Tau protein which induces depolymeri-
zation of microtubules, leading to the formation of neu-
rofibrillary tangles and destabilization of the neuronal
processes. Microtubules are cytoskeletal components
that are involved in the maintenance of neuronal mor-
phology and axon and dendrite formation processes
[168, 169].
Thiazolidinones (TZD) were the first ATP-non-
competitive inhibitors (Table 1) with a potential to
originate drugs for the treatment of AD [170]. How-
ever, other compounds such as bis-indole [171], aniline
[172], maleimides, kenpaullone [173] indirubin [174]
and hymenialdisin [175] have been described as inhibi-
tors of GSK-3β.
The study by Hu et al., [161] corroborated the hy-
pothesis that hyperphosphorylation of the GKS-3 is
involved in the neurodegeneration-induced Abeta pep-
tide. In this study, SB216763 (3-[2,4-dichlorophenyl]-
4-[1-methyl-1H-indol-3-yl] -1H-pyrrole-2,5-dione)
inhibits the GSK-3 enzyme. The SB216763 is a highly
selective inhibitor that possesses cell permeability and
acts by competing with ATP binding site. The destabi-
lization of microtubules induced by rotenone, a toxin
that inhibits the mitochondrial complex I, was attenu-
ated by using the GSK-3β inhibitor SB216763 in hu-
man neuroblastoma cells SH-SY5Y [167]. According
to Wang et al., [176], the SB216763 is a potent imag-
ing agent that can be used in positron emission tomo-
graphy (PET) of GSK-3.
To determine the relationship via PI3-K/Akt/GSK-
3β with the NMDA receptor signaling pathway in glu-
tamate-induced excitotoxicity, the SB415286 inhibitor
was used (pre-treatment) in primary cultures of cere-
bellar granular neurons in rats and it was observed that
this compound blocks the glutamate induced apoptosis
[144]. Moreover, mice pretreated with inhibitors
SB216763 (0.6 mg/kg) and SB415286 (1 mg/kg) for 3
days before intracranial radiation of a single dose of 7
grays (Gy), improved their performance in the Morris
water maze platform [177].
Lithium was the first inhibitor of GSK-3 discovered
and presents two possible mechanisms of action: it may
cause deprivation of potassium or inhibit the enzyme
competing with magnesium ions (Mg2+). Lithium, be-
sides reducing hyperphosphorylation of the Tau pro-
tein, is capable of reducing the production of Abeta
peptide [178-180]. Noble et al., [181] observed a sig-
nificant reduction in GSK-3 activity and Tau protein
phosphorylation after administering intraperitoneally
lithium for 30 days in transgenic mice that overexpress
the Tau human protein.
Lithium and valproate are inhibitors of the enzyme
GSK-3β, and reduced the phosphorylation of the Tau
protein in animal models, according to Tariot and
Aisen [182]. However, divalproex sodium has acceler-
ated the loss of brain volume after one year of treat-
ment, in addition to a greater cognitive impairment ac-
cording to Fleisher et al., [183].
In transgenic mice treated with a peptide inhibitor of
GSK-3, the mts-L803, it was observed the inhibition of
Abeta peptide accumulation and improvement in cogni-
tive function [184]. According to Berg et al., [185],
piperazinyl sulfonamide analogs inhibit GSK-3β, be-
sides having good solubility and permeability through
the blood brain barrier and Caco-2 assays.
In a study by Coffman et al., [186], derivatives of 6-
amino-4-(pyrimidin-4-yl)pyridone showed inhibitory
activity against the GSK-3β enzyme in a cellular en-
zyme inhibition assay. Also, the 2-(2-phenylmorpholin-
4-yl)pyrimidin-4(3H)-ones decreased phosphorylation
of the Tau protein in mice when administered orally, by
inhibiting GSK-3β [169].
The 1,3,4-oxadiazole derivatives are potent and
highly selective inhibitors of GSK-3β, three of these
compounds (2-methyl-5-{3-[4-(methylsulfonyl)-, 2-
methyl-5-{3-[4-(methylsulfinyl)-, and 2-methyl-5-{3-
[4-(methylsulfonyl)-(3)-phenyl]-1-benzofuran-5-yl}-
1,3,4-oxadiazoles (MMBO), IC50 = 66, 35 and 42 nM,
respectively) showed good pharmacokinetic profiles
and high absorptions by the blood brain barrier. The
MMBO showed high selectivity for GSK-3 in inhibit-
ing the phosphorylation of the Tau protein in cultured
primary neural cells and normal rat brain, showing to
be a good drug candidate for the treatment of AD [187-
190].
Two quinolone derivatives (IC50 = 35 and 158 nM)
had inhibitory activity against GSK-3β and neuropro-
tective action against the injury caused by the produc-
tion of Abeta peptide in MC65 cells [191]. The prolyl
isomerase Pin1, an enzyme that controls the phos-
phorylation of proteins, inhibits GSK-3β activity and
decreased the PPA levels, which demonstrates a new
protection mechanism against AD [192].
12 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
Compounds C-7a and C-7b (120-130 nM) were able
to interfere with the neuronal death-induced accumula-
tion of Abeta peptides and to inhibit the phosphoryla-
tion of the Tau protein (pThr231, pSer396, pThr181
and pSer202) in vitro. Furthermore, in vivo, 50 mg/kg
of C-7a decreased the phosphorylation of the Tau pro-
tein (Ser202) in the hippocampus of the brains of mice.
The C-7a may be considered a disease-modifying agent
and a potent candidate for the treatment of AD [193].
According Uehara et al., [194] pyrimidones showed
inhibitory activity against GSK-3β and good perme-
ability in mouse brains and reduction of the phosphory-
lation of the Tau protein. According Giacobini and
Gold [195] some GSK-3β inhibitors are in clinical
phase trials, such as Np-1003, developed by the
Noscira S.A., SAN-161 (Sanoimmune) and an inhibitor
of the aggregation of the Tau protein, methylthion-
inium chloride (TRx0237) studied at the University of
Aberdeen and TauRx Therapeutics.
6.5. Complementary Treatments Applied to AD
Regarding the pharmacological treatment of AD,
several authors state that, in addition to therapies al-
ready described in this article, there are other ap-
proaches that can collaborate in the process of stabili-
zation of the disease, such as the use of anti-
inflammatory, antioxidant, estrogenic replacement and
vaccine, among others [196-198].
The anti-inflammatory drugs (Table 1) act by reduc-
ing the inflammatory response in the brain tissue, as in
the studies conducted by Lim et al., (2001) [199],
which suggested that indomethacin and ibuprofen
would be able to reduce Abeta formation. However,
prolonged use can cause many unwanted side effects
such as kidney and stomach problems [200]. Other ex-
amples of anti-inflammatory drugs are: curcumin, ca-
pable of modulating eicosanoid biosynthesis and inhib-
iting COX-1, COX-2 and LOX; Ginkgolide B, which is
an antagonist of platelet activating factor; magnolol; Α-
pinene; Beta-pinene; carvacrol; luteolin; Rosmarinic
acid; resveratrol; and apocynin, which also act as in-
hibitors of COX. Furthermore, the compound 4'-O-
methylhonokiol (MH) has demonstrated an important
inhibitory capacity on the induction of amyloidogenesis
through anti-inflammatory mechanisms. It inhibits the
activation of astrocytes in brain tissue and several other
signaling cascades related to inflammation and oxida-
tive stress. Thus, all these activities favor the inhibition
of the inflammatory processes associated with the
physiology of AD and reduce the deleterious conse-
quences of this disease [133, 196, 200, 201].
Antioxidants prevent free radical formation, reduc-
ing oxidative stress on the cells and thus assisting in the
treatment of AD, exerting a probable neuroprotective
effect. The vitamin E demonstrated benefits on AD,
slowing the natural course of the disease [133]. Other
examples of antioxidants are resveratrol, apocynin
[196], and some flavonoids such as quercetin and
myricetin, which through their known antioxidant ac-
tivity prevent the inactivation of acetylcholine recep-
tors [74]. Similarly, the estrogen-replacement therapy
also acts through neuroprotective mechanisms in the
prevention of AD. Estrogens exert effects on various
receptors of the neuronal surface, promoting the release
of neurotransmitters and increasing the cerebral blood
flow. In addition, there are cases of estrogen reducing
the neurotoxicity promoted by Abeta [165]. Regarding
the anti-Alzheimer vaccine AT-1792, it was developed
to act on amyloid plaques but showed significant ad-
verse effects leading to a discontinuation of its use
[176].
CONCLUSION
The multifactorial character of AD's pathogenic
mechanism hinders the development of fully effective
drugs for its therapy. Thus, several researchers con-
tinue in the search for new drug candidates for the
treatment of AD. Nevertheless, all the drugs available
so far act only on the symptoms, which mean that these
treatments are often found to be unsatisfactory in view
of a permanent stabilization of the disease after the di-
agnosis. In addition, maintenance of the treatment from
the patient himself or caregiver is often difficult, since
this type of disease often requires drug combinations to
achieve better clinical outcomes. It is in this aspect that
the early diagnosis facilitates the maintenance of the
memory and cognitive functions of these patients. The
use of various drugs and drug candidates discussed in
this article may help to improve the prospects of the
anti-Alzheimer's therapy.
CONFLICT OF INTEREST
The authors confirm that this article content has no
conflict of interest.
ACKNOWLEDGEMENTS
We gratefully acknowledge the support provided by
the Postgraduate Program in Pharmaceutical Sciences
(UNIFAP), Laboratory of Modeling and Computational
Chemistry of Federal University of Amapá (UNIFAP)
and Computational Laboratory of Pharmaceutical
Chemistry of Faculty of Pharmaceutical Sciences of
Ribeirão Preto in University of São Paulo.
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 13
REFERENCES
[1] Bonda, D.J.; Wang, X.; Perry, G.; Nunomura, A.; Tabaton,
M.; Zhu, X.; Smith, M. A. Oxidative stress in Alzheimer
disease: a possibility for prevention. Neuropharmacology,
2010, 59, 290-294.
[2] Easton, A.; Sankaranarayanan, S.; Tanghe, A.; Terwel, D.;
Lin, A.X.; Hoque, N.; Bourin, C.; Gu, H.; Ahlijanian, M.;
Brislow, L. Effects of subchronic donepezil on brain Abeta
and cognition in a mouse model of Alzheimer’s disease.
Psychopharmacology (Berl), 2013, 230(2), 279-289.
[3] Lu, C.; Guo, Y.; Yan, J.; Luo, Z.; Luo, H.; Yan, M.; Huang,
L.; Li, X. Design, Synthesis, and Evaluation of Multitarget-
Directed Resveratrol Derivatives for the Treatment of Alz-
heimer’s Disease. J. Med. Chem., 2013, 56(14), 5843-5859.
[4] Richardson, C.; Gard, P.R.; Klugman, A.; Isaac, M.; Tabet,
N. Blood pro-inflammatory cytokines in Alzheimer’s dis-
ease in relation to the use of acetylcholinesterase inhibitors.
Int. J. G eriatr. Psychiatry, 2013, 28, 1312-1317.
[5] Reitz, C.; Mayeux, R. Alzheimer disease: Epidemiology,
diagnostic criteria, risk factors and biomarkers. Biochem.
Pharmacol., 2014, 88(4), p. 640651.
[6] World Alzheimer Report 2015. alz.co.uk/research/world-
report-2015 (Accessed May 02, 2016).
[7] World Alzheimer Report 2010.www.alz.co.uk/research/
files/WorldAlzheimerReport-2010.pdf (Accessed May 02,
2016)
[8] Manual Diagnostico e Estatistico de Transtornos Mentais-
DMS-5. 5rd ed.: Artmed: Porto Alegre, 2014.
[9] Inouye, K.; Pedrazzani, E.S.; Pavarini, S.C.I. Influencia da
doenca de Alzheimer na percepcao de qualidade de vida do
idoso. Rev. Esc. Enferm. USP, 2010, 44(4), 1093-1099.
[10] Weiner, M.W.; Friedl, K.E.; Pacifico, A.; Chapman, J.C.;
Jaffee, M.S.,;Little, D.M.; Manley, G.T.; McKee, A.; Peter-
sen, R.C.; Pitman, R.K.; Yaffe, K.; Zetterberq, H.; Obana,
R.; Bain, LJ.; Carrillo, M.C. Military risk factors for Alz-
heimer’s disease. Alzheimers Dement, 2013, 9, 445451.
[11] Fischer H.U.S. military casualty statistics: Operation New
Dawn, Operation Iraqi Freedom, and Operation Enduring
Freedom.Washington DC: Congressional Research Service;
2010. p. 18.
[12] Plassman, B.L.; Havlik, R.J.; Steffens, D.C.; Helms, M.J.;
Newman, T.N.; Drosdick, D.; Phillips, C.; Gau, B.A.;
Welsh-Bohmer, K.A.; Burke, J.R.; Guralnik, J.M.; Breitner,
J.C. Documented head injury in early adulthood and risk of
Alzheimer’s disease and other d ementias. Neurology, 2000;
55, 11581166.
[13] Martland, H. Dementia pugilistica. JAMA, 1928, 91, 1103
1107.
[14] Langlois J.A.; Rutland-Brown, W.; Wald, M.M. The epi-
demiology and impact of traumatic brain injury: a brief
overview. J. Head Trauma Rehabil., 2006, 21, 375378
[15] Nowinski C. Head games: football’s concussion crisis from
the NFL to youth leagues. East Bridgewater, MA: Drum-
mond Publishing Group; 2006.
[16] Thurman, D.J.; Branche, C.M.; Sniezek, J.E. The epidemi-
ology of sportsrelated traumatic brain injuries in the United
States: recent developments. J Head Trauma Rehabil.,
1998, 13, 18.
[17] Instituto Brasileiro de Geografia e Estatistica (IBGE). Sin-
tese de Indicadores So ciais Uma Analise das Condicoes de
Vida da Populacao Brasileira (2012). biblio-
teca.ibge.gov.br/visualizacao/livros/liv62715.pdf (Accessed
May 02, 2016).
[18] Alzheimermed. Epidemiologia da Doenca de Alzheimer.
alzheimermed.com.br/conceitos/epidemiologia (Accessed
May 02, 2016).
[19] Herrera Jr, E.; Caramelli, P.; Silveira, A.S.; Nitrini, R. Epi-
demiologic survey of dementia in a community-dwelling
Brazilian population. Alzheimer Dis. Assoc. Disord., 2002,
16(2), 103-108.
[20] Ferri, C.P. Population ageing in Latin America: dementia
and related disorders. Rev. Bras. Psiquiatr., 2012, 34(4),
371-372.
[21] Forestier, A.; Douki, T.; De Rosa, V.; Beal, D.; Rachidi, W.
Combination of Aβ Secretion and Oxidative Stress in an
Alzheimer-Like Cell Line Leads to the Over-Expression of
the Nucleotide Excision Repair Proteins DDB2 and XPC.
Int. J. Mol. Sci., 2015, 16, 1742217444.
[22] Liu, J.; Liu, Z.; Zhang, Y.; Yin, F. A Novel Antagonistic
Role of Natural Compound Icariin on Neurotoxicity of
Amyloid β Peptide. Indian J. Med. Res., 2015, 142, 190
195.
[23] Pereira, P.M.C.M. Doença de Alzheimer: Perspetivas de
Tratamento, Universidade da Beira Interior, 2013.
[24] Alves, L.; Correia, A.S.A.; Miguel, R.; Alegria, P.;
Bugalho, P. Alzheimer’s Disease: A Clinical Practice-
Oriented Review. Front. Neurol., 2012, 3, 120.
[25] Forestier, A.; Douki, T.; De Rosa, V.; Beal, D.; Rachidi, W.
Combination of Aβ Secretion and Oxidative Stress in an
Alzheimer-Like Cell Line Leads to the Over-Expression of
the Nucleotide Excision Repair Proteins DDB2 and XPC.
Int. J. Mol. Sci., 2015, 16, 1742217444.
[26] Liu, J.; Liu, Z.; Zhang, Y.; Yin, F. A Novel Antagonistic
Role of Natural Compound Icariin on Neurotoxicity of
Amyloid β Peptide. Indian J Med Res, 2015, 142, 190195.
[27] Stancu, I.; Vasconcelos, B.; Terwel, D.; Dewachter, I. Mod-
els of β-Amyloid Induced Tau-Pathology : The Long and
“Folder” Road to Understand the Mechan ism. Mol. Neu-
rodegener., 2014, 9, 114.
[28] Holtzman, D.M.; John, C.M.; Goate, A. Alzheimer’s Dis-
ease : The Challenge of the Second Century. Sci Transl
Med., 2012, 3, 135.
[29] Perl, D.P. Neuropathology of Alzheimer’s Disease. Mt.
Sinai. J. Med., 2010, 77, 3242.
[30] Hendrie, H.C.; Hake, A.; Lane, K.; Purnell, C.; Unverzagt,
F.; Smith-gamble, V.; Murrell, J.; Ogunniyi, A.; Baiyewu,
O.; Callahan, C.; Saykin, A.; Taylor, S.; Hall, K.; Gao, S.
Statin Use, Incident Dementia and Alzheimer Disease in
Elderly African Americans. Ethn. e Dis., 2015, 25, 345
354.
[31] Zhu, L.; Zhong, M.; Elder, G.A.; Sano, M.; Holtzman,
D.M.; Gandy, S.; Cardozo, C.; Haroutunian, V.; Robakis,
N.K.; Cai, D. Phospholipid Dysregulation Contributes to
ApoE4- Associated Cognitive Deficits in Alzheimer’s Dis-
ease Pathogenesis. Proc. Natl. Acad. Sci. 2015, 112, 11965
11970.
[32] Kunz, L.; Schroder, T.N.; Lee, H.; Montag, C.; Lachmann,
B.; Sariyska, R.; Reuter, M.; Stirnberg, R.; Stocker, T.;
Messing-floeter, P.C.; Fell, J.; Doeller, C.F.; Axmacher, N.
Reduced Grid-Cell-like Representations in Adults at Ge-
netic Risk for Alzheimer’s Diseade. Science. 2015, 350,
430434.
[33] Goedert, M.; Spillantini, M.G.; Crowther, R.A. A Brief
History of Tau. Clin. Chem., 2015, 61(11), 14171418.
[34] Prado, D.; Cardoso, I..L. Apolipoproteina E e Doenca de
Alzheimer. Rev. Neurocienc. 2013, 21(1), 118125.
[35] Cabezas-opazo, F.A.; Vergara-pulgar, K.; Perez, M.J.; Jara,
C.; Osorio-fuentealba, C.; Quintanilla, R.A. Mitochondrial
Dysfunction Contributes to the Pathogenesis of Alzheimer’s
Disease. Oxid. Med. Cell. Longev., 2015, 2015, 1-12.
[36] Li, J.; Kanekiyo, T.; Shinohara, M.; Zhang, Y.; LaDu, M.J.;
Xu, H.; Bu, G. Differential Regulation of Amyloid-β Endo-
cytic Trafficking and Lysosomal Degradation by Apolipo-
14 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
protein E Isoforms. J. Biol. Chem., 2012, 287, 44593
44601.
[37] Aguayo, L. G. H.; Oprazo, C. R. M.; Parodi, L. R. Real
time electrophysiological testing of agents of interest for
treatment of amyloid-type diseases. Unit States Patent,
2013.
[38] Bagyinszky, E.; Youn, Y.C.; An, S.S.A.; Kim, S.Y. The
genetics of Alzheimer’s disease. Clin. Interv. Aging. 2014,
9, 535551.
[39] Arango D.; Cruts, M.; Torres, O.; Backhovens, H.; Serrano,
M.L.; Villareal, E.; Montanes, P.; Matallana, D.; Cano, C.;
Van Broeckhoven, C.; Jacquier M. Systematic genetic study
of Alzheimer disease in Latin America: mutation frequen-
cies of the amyloid beta precursor protein and presenilin
genes in Colombia. Am. J. Med. Genet. 2001, 103(2):138-
143.
[40] Modarresi, F.; Faghihi, M.A.; Patel, N.S.; Sahagan, B.G.;
Wahlestedt, C.; Lopez-Toledano, M.A. Knockdown of
BACE1-AS Nonprotein-Coding Transcript Modulates Beta-
Amyloid-Related Hippocampal Neurogenesis. Int. J. Alz-
heimers Dis. 2011, 111.
[41] Friedman, L.G.; Qureshi, Y.H.; Yu, W.H. Promoting
Autophagic Clearance : Viable Therapeutic Targets in Alz-
heimer’s Disease. Neurotherapeutics, 2015, 94108.
[42] Rogaeva, E.; Tandon, A.; St George-Hyslop, P. H. The
solved and unsolved mysteries of the genetics of early-onset
Alzheimer’s disease. Neuro. Molec. Medicine, 2002, 2, 33-
42.
[43] Cai, Y.; An, S. S.; Kim, S. Mutations in presenilin 2 and its
implications in Alzheimer’s disease and other dementia-
associated disorders. Clin. Interv. aging, 2015, 10, 1163-
1172.
[44] Suh, Y.; Checler, F. Amyloid Precursor Protein, Presenilins,
and α-Synuclein: Molecular Pathogenesis and Pharmacol-
ogical Applications in Alzheimer’s Disease. Pharmacol.
Rev., 2002, 54, 469-525.
[45] Lobo, A.; Launer, L. J.; Fratiglione, L. Et al. Prevalence of
dementia and major subtype in Europe: a collaborative
study of population-based cohorts. Neurology. 2000, 11,
S4-S9.
[46] Golde, T. E. Disease modifying therapy for AD. J. Neuro-
chem. 2006, 3, 689-707.
[47] Rang, H. P.; Dale, M.M.; Ritter, J.M.; Flower, R.J.;
Henderson, G. Farmacologia. 7rd ed. Rio de Janeiro: El-
sevier, 2011.
[48] Vasques, J. F. Expressao e processamento da proteina pre-
cursora de amiloide no coliculo superior de ratos subm eti-
dos a enucleacao monocular. Thesis, 2012, 123.
[49] Kreutz, F. Efeito do peptide beta-amilpoide sobre a bios-
sitese de gangliosideos e avalialcao da atividade neuropro-
tetora do GM1. Thesis, 2010, 93.
[50] Ritter, A.; Cummings, J. Fluid biomarkers in clinical trials
of Alzheimer’s disease therapeutics. Front. Neurol., 2015,
6,186.
[51] Furazo Junior, G; Nivel de atividade fisica, estado nutri-
cional e niceis de CABE1 e BACE2 em idosos neurologi-
camente saudpaveis e com doenca de Alzheimer. Thesis,
2015, 103.
[52] Salim, O.; Abrahin, C. Influencia Das Proteinas Beta Ami-
loide e Tau Na Doenca de Alzheimer. Facen/Famene, 2012,
10(1), 5760.
[53] Hardy, J. A.; Higgins, G. A. Alzheimer’s disease: the amy-
loid cascade hypothesis. Science, 1992, 256, 184-185.
[54] De Falco, A.; Cukierman, D. S.; Hauser-Davis, R. A.; Rey,
N. A. Doenca de Alzheimer: hipoteses etiologicas e per-
spectivas de tratamento. Quím. Nova, 2016, 39, 63-80.
[55] Marteli, A.; Marteli, F.P. Alteracoes Cerebrais e Analise
Histopatologica dos Emaranhados Neurofibrilares na
Doenca de Alzheimer. Uniciências, 2014, 18(1), 45-50.
[56] Lacor, P. N.; Buniel, M. C.; Chang, L.; Fernandez, S. J.;
Gong, Y.; Viola, K. L.; Lambert, M. P.; Velasco, P. T.;
Bigio, E. H.; Finch, C. E.; Krafft, G.A.; Klein, W. L. Syn-
aptic targeting by Alzheimer’s related amyloid beta oli-
gomers. J. Neurosci., 2004, 24, 10919-10200.
[57] Jonsson, T.; Stefansson, H.; Steinberg, S.; Jonsdottir, I.;
Jonsson, P. V.; Snaedal, J.; Bjornsson, S.; Huttenloch er, J.;
Levey, A.I.; Lah, J.J.; Rujescu, D.; Hampel, H.; Giegling,
I.; Andreassen, O.A.; Engedal, K.; Ulstein, I.; Djurovic, S.;
Ibrahim-Verbaas, C.; Hofman, A.; Ikram, M.A.; van Duijn,
C.M.; Thorsteinsdottir, U.; Kong, A.; Stefansson, K. Vari-
ant of TREM2 Associated with the Risk of Alzheimer’s
Disease. N. Engl. J. Med., 2013, 368, 107116.
[58] Bhaskar, K.; Konerth, M.; Kokiko-Cochran, O.N.; Cardona,
A.; Ransohoff, R.M.; Lamb, B.T. Regulation of Tau Pa-
thology by the Microglial Fractalkine Receptor. NIH Public
Access, 2011, 68, 1931.
[59] Sy, M.; Kitazawa, M.; Medeiros, R.; Whitman, L.; Cheng,
D.; Lane, T.E.; LaFerla, F.M. Inflammation Induced by In-
fection Potentiates Tau Pathological Features in Transgenic
Mice. Am. J. Pathol., 2011, 178, 28112822.
[60] Christensen, D.D. Changing the course of Alzheimer's dis-
ease: anti-amyloid disease-modifying treatments on the ho-
rizon; Prim. Care Companion J. Clin. Psychiatry, 2007,
9(1), 32-41.
[61] Benevento, C.E. Disfunção mitocondrial induzida por pep-
tídeos beta-amilóide, Universidade Estadual de Campinas,
2011.
[62] Schwab, C.; McGeer, P. L. Inflamatory aspects of Alz-
heimer Disease and other neurodegenerative desorders. J.
Alzheimer Dis., 2008, 13, 359-369.
[63] Barnham, K.J.; Masters, C.L.; Bush, A.I. Neurodegenera-
tive diseases and oxidative stress. Nat. rev. Drug Discov.
2004, 3, 205-214.
[64] Bartus, R. T.; Dean, R. L.; Beer, B.; Lippa, A. S. The cho-
linergic hypothesis of geriatris memory dysfunction. Sci-
ence, 1982, 217, 408-414.
[65] Racchi, M.; Mazzucchelli, M.; Porrelo, E.; Lanni, C.; Go-
voni, S. Acetylcholinesterase inhibitors: novel activities of
old molecules. Pharmacol. Res., 2004, 50, 441-450.
[66] Lunardi, P. S. Efeito da Huperzina-A sobre a secrecao de
S100B em cultura primaria de astrocitos e fatias hipocam-
pais. Thesis, 2010, 44.
[67] Bartus, R. T.; Emerich, D. F. Cholinergic markes in Alz-
heimer’s disease. JAMA, 1999, 23, 2208-2209.
[68] Kasa, P.; Rakonczay, Z.; Gulya, K. The cholinergic system
in Alzheimer’s disease. Prog. Neurobiol.., 1997, 52, 511-
535.
[69] Cummings, J. Alzheimer’s Disease. New Engl. J. Med.,
2004, 351, 56-67.
[70] Aguera Morales, E.; Tunez Finana, I. Demencia. Medicine
(Baltimore). 2011, 10 (76), 51235128.
[71] Franco Macias, E.; Suarez Gonzalez, A.; Bustamante
Rangel, A.; Gil Neciga, E. Enfermedad de Alzheimer.
Medicine (Baltimore). 2011, 10 (76), 51295137.
[72] American Psychiatric Association. Diagnostic and Statisti-
cal Manual of Mental Disorders, 4th. Diagnostic Stat. Man.
Ment. Disord. 4th Ed. TR. 2000.
[73] Hampel, H.; Prvulovic, D.; Teipel, S.; Jessen, F.; Luckhaus,
C.; Frolich, L.; Riepe, M.W.; Dodel, R.; Leyhe, T.;
Bertram, L.; Hoffmann, W.; Faltraco, F. The Future of Alz-
heimer’s Disease: The next 10 Years. Prog. Neurobiol.
2011, 95 (4), 718728.
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 15
[74] De Melo, G.O.; Costa, S.S. Produtos Naturais para o
Tratamento da Doenca de Alzheimer: Promessa e Desafio.
Rev. Fitos., 2005, 1 (2), 4147.
[75] Ghanemi, A. Alzheimer’s Disease Therapies: Selected Ad-
vances and Future Perspectives. Alexandria J. Med. 2015,
51 (1), 13.
[76] Seidl, C. Pesquisa de substâncias naturais inibidoras da
acetilcolinesterase. Universidade Federal do Parana, 2010.
[77] Seima M.D.; Lenardt, M.H. A sobrecarga do cuidador fa-
miliar de idoso com Alzheimer. Textos Contextos, 2011,
10(2), 338-398.
[78] Lindeboom J.; Weinstein, H. Neuropsychology of cognitive
ageing, minimal cognitive impairment, Alzheimer's disease,
and vascular cognitiv e impairment. Eur J Pharmacol.,
2004; 490(1-3), 83-86.
[79] Ballard, C.; Gauthier, S.; Corbett, A.; Brayne, C.; Aarsland,
D.; Jones, E. Alzheimer’s Disease. Lancet, 2011,
377(9770), 10191031.
[80] Inouye, K.; Oliveira, G.H.D.E. Avaliacao Critica Do
Tratamento Farmacologico. Infarma, 2004, 15, 8084.
[81] Gross, R.G.; Grossman, M. Update on Apraxia. Curr. Neu-
rol. Neurosci. Rep., 2008, 8 (6), 490496.
[82] Bottino, C.M.C.; Carvalho, I.A.M.; Alvarez, A.M.M.A.;
Avila, R.; Zukauskas, P. R.; Bustamante, S.E.Z.; Andrade,
F.C.; Hototian, S.R.; Saffi, F.; Camargo, C.H.P. Reabilita-
cao Cognitiva Em Pacientes Com Doenca de Alzheimer:
Relato de Trabalho Em Equipe Multidisciplinar. Arq.
Neuropsiquiatr., 2002, 60(1), 7079.
[83] Petronilho, E.D.C.; Pinto, A.C.; Villar, J.D.F. Acetil-
colinesterase: Alzheimer e Guerra Quimica. Ciência e Tec-
nol., 2011, 314.
[84] Camargo, I.E. Doenca de Alzheimer - um testemunho pela
preservacao da vida. Ed. AGE: Porto Alegre, 2003.
[85] Moreaud, O.; David, D.; Brutti-Mairesse, M.P.; Debray,
M.; Memin, A. [Aphasia in elderly patients]. Psychol Neu-
ropsychiatr Vieil. 2010, 8(1), 43-51.
[86] Helito, A.; Kauffman, P. Saude: Entendendo as doencas, a
enciclopedia medica da familia. Nobel: Sao Paulo, 2003.
[87] Aging, N.I. Alzheimer’ Disease: Unraveling the Mystery.
Nat. Inst. Aging. 2008, 5-7.
[88] Rosenberg, P.B.;, Nowrangi, M.A.; Lyketsos, C.G. Neuro-
psychiatric symptoms in Alzheimer’s disease: what might
be associated brain circuits? Mol. Aspects. Med., 2015,
43(44), 111.
[89] NIH Instituto Nacional sobre el Envejecimiento. Guia Para
Quienes Cuidan a Personas Con Alzheimer. 2006.
[90] Hughes, C.P.; Berg, L. A new clinical scale for the staging
of dementia. Br. J. Psychia try, 1982, 140(6), 566572.
[91] Haupt, M.; Kurz, A.; Janner, M. A 2-year follow-up of be-
havioural and psychological symptoms in Alzheimer’s dis-
ease. Dement. Geriatr. Cogn. Disord.,. 2000, 11(3), 147-
152.
[92] Diamond, J. Report on Alzheimer’s Disease and current
research. Alzheim er Society of Canada: Toronto, 2008.
[93] Mckhann, G. Clinical diagnosis of Alzheimer’s disease:
report of the NINCDS-ADRDA work group under the aus-
pices of department of health and human services task force
on Alzheimer’s disease. Neurology, 1984, 34, 939-944.
[94] Folstein, M.F.; Folstein, S.E.; Mchugh, P.R. Mini-mental
state: a practical method for grading the cognitive state of
patients for the clinician. J. Psychiatr. Res., 1975, 12, 189-
198.
[95] Almeida, O.P. Mini-exame do estado mental e o diagnos-
tico de demencia no Brasil. Arq. Neuropsiquiatr., 1998,
56(3B), 605-612.
[96] Nitrini, R.; Caramelli, P .; Bottino, C.M.C.; Damasceno,
B.P.; Brucki, S.M.D.; Anghinah, R. Diagnostico de doenca
de Alzheimer no Brasil. Arq. Neuropsiquiatr, 2005, 63(3-
A), 720-727.
[97] Burns, A.; Lawlor, B.; Craig, S. Blessed Dementia Scale
(incorporating the Information-Memory Concentration
(ICM) Test and the Dementia Scale. In: Assessment Scales
in Old Age Psychiatry. Martin Dunitz: London, 1999; pp.
40-41.
[98] Burns, A.; Lawlor, B.; Craig, S. Cognitive Abilities Screen-
ing Instrument (CASI). In: Assessment Scales in Old Age
Psychiatry. Martin Dunitz:London, 1999; pp. 63.
[99] Welsh, K.; Butters, N.; Hughes, J.; Mohs, R.; Heyman, A.
Detection of abnormal memory decline in mild cases of
Alzheimer’s disease using CERAD neuropsychological
measures. Arch. Neurol., 1991, 48(3), 278-281.
[100] Rey, A. L' exam clinique en psychologie. Presses Un iversi-
taire de France: Paris, 1964.
[101] Weschler, D. Weschler Memory Scale - revised manual.
Psychological Corporation: New York, 1987.
[102] Bertolucci, P.H.F.; Okamoto, I.H.; Brucki, S.M.D .; Siviero,
M.O.; Neto, J.T.; Ramos, L.R. Applicability of the CERAD
neuropsychological battery to Brazilian elderly. Arq. Neu-
ropsiquiatr., 2001, 59(3A), 532-536.
[103] Strub, R.L.; Black, F.W. The mental status examination in
neurology. 2nd ed.; F.A. Davis Company: Philadelphia,
1985.
[104] Lezak, M.D. Neuropsychological assessment. Oxford
University Press: New York, 1995.
[105] Rosen, W.G.; Mohs, R.C.; Davis, K.L. A new rating scale
for A lzheimer’s disease. The American Journal of Psychia-
try, 1984, 141(11), 1356-1364.
[106] Kaplan, E.; Goodglass, H.; Weintraub, S. Boston Naming
Test. Lea & Febiger: Philadelphia, 1983.
[107] Ostrosky-Solis, F.; Ardila, A.; Rosselli, M. NEUROPSI: a
brief neuropsychoogical test battery in Spanish with norms
by age and educational level. J. Int. Neuropsychol. Soc.,
1999, 5(5), 413-433.
[108] Roth, M.; Tym, E.; Mountjoy, C.Q.; Huppert, F.A.; Hen-
drie, H.; Verma, S.; Goddard, R. CAMDEX - A standard-
ized instrument for the diagnosis of mental disorder in the
elderly with special reference to the early detection of de-
mentia. Br. J. Psychiatry., 1986, 149(6), 698-709.
[109] Bottino, C.M.C.; Stoppe Jr, A.; Scalco, A.Z.; Ferreira,
R.C.R.; Hototian, S.R.; Scalco, M.Z. Validade e confiabili-
dade da versao brasileira do CAMDEX. Arq. Neurop-
siquiatr., 2001, 59(3), 20.
[110] Shulman, K.I. Clock-drawing: is it the ideal cognitive
screening test? International Journal of Geriatric Psychia-
try, 2000, 15(6), 548-561.
[111] Petersen, R.C.; Smith, G.E.; Waring, S.C.; Ivnik, R.J.; Tan-
galos, E.G.; Kokmen, E. Mild cognitive impairment: clini-
cal characterization and outcome. Archi. N eurol., 1999,
56(3), 303-308.
[112] Winblad, B.; Palmer, K.; Kivipelto, M.; Jelic, V.; Fratigli-
oni, L.; Wahlund, L.O.; Nordberg, A.; Backman, L.; Albert,
M.; Almkvist, O.; Arai, H.; Basun, H.; Blennow, K.; de
Leon, M.; DeCarli, C.; Erkinjuntti, T.; Giacobini, E.; Graff,
C.; Hardy, J.; Jack, C.; Jorm, A.; Ritchie, K.; Van Duijn, C.;
Visser, P.; Petersen, R.C. Mild cognitive impairment-
beyond controversies, towards a consensus: report of the In-
ternational Working Group on Mild Cognitive Impairment.
J. Intern. Med., 2004, 256(3), 240-246.
[113] Caramelli, P.; Teixeira, A.L.; Buchpiguel, C.A.; Lee, H.W.;
Livramento, J.A.; Fernandez, L.L.; Anghinah, R. Grupo
para Recomendacoes em DA e Demencia Vascular da Aca-
demia Brasileira de Neurologia. Diagnostico de doenca de
Alzheimer no Brasil Exames complementares. Dement.
Neuropsychol., 2011, 5(3), 11-19.
16 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
[114] Nitrini, R., Caramelli, P., Bottino, C.M., Damasceno, B.P.,
Brucki, S.M., Anghinah, R.; Academia Brasileira de Neu-
rologia. Diagnostico de doenca de Alzheimer no Brasil: cri-
terios diagnosticos e exames complementares. Arq Neurop-
siquiatr., 2005; 63: 713-719.
[115] Bouwman, F.H.; Schoonenboom, S.N.; Van der Flier,
W.M.; Van Elk, E.J.; Kok, A.; Barkhof, F.; Blankenstein,
M.A.; Scheltens P. CSF biomarkers and medial temporal
lobe atrophy predict dementia in mild cognitive impairment.
Neurobiol. Aging, 2007, 28(7), 1070-1074.
[116] Hansson, O.; Zetterberg, H.; Buchhave, P.; Londos, E.;
Blennow, K.; Minthon, L. Association between CSF bio-
markers and incipient Alzheimer’ s disease in patients with
mild cognitive impairment: a follow-up study. Lancet Neu-
rol., 2006, 5(3), 228-234.
[117] Herukka, S.K.; Helisalmi, S.; Hallikainen, M.; Tervo, S.;
Soininen, H.; Pirttila T. CSF Aβ42, Tau and phosphorylated
Tau, APOE e4 allele and MCI type in progressive MCI.
Neurobiol. Aging, 2007, 28(7), 507-514.
[118] Fagan, A.M., Mintun, M.A, Mach, R.H, et al. Inverse rela-
tion between in vivo amyloid imaging load and cerebrospi-
nal fluid Abeta42 in humans. Ann Neurol., 2006, 59, 512-
519.
[119] Costa, D.C.; Oliveira, J.M.A.P.; Bressan, R.A. PET e
SPECT em neurologia e psiquiatria: do basico as aplicacoes
clinicas. Rev. Bras. Psiquiatr., 2001, 23(Suppl. 1), 4-5.
[120] Grasso, M.; Piscopo, P.; Confaloni, A.; Denti, M.A. Circu-
lating miRNAs as biomarkers for n eurodegenerative disor-
ders. Molecules, 2014, 19, 68916910.
[121] Charlotte, D.A.B.; Wim, M.C.D.; Sebastien, S.H.U.B. Mi-
croRNAs in Alzheim er's disease. Neurobiol. Dis., 2012,
46(2), 285290.
[122] Almeida, J.R. Estudos de modelagem molecular e relacao
estrutura-atividade da acetilcolinesterase e inibidores em
Mal de Alzheimer. Dissertacao, Universidade d e Sao Paulo:
Ribeirao Preto, 2011.
[123] Cruz, D. Estudos de QSAR 2D e 3D para derivados de ami-
noimidazois, aminohidantoinas e aminipiridinas com ac-
tividade inibitoria sobre a enzima beta-secretase humana.
Dissertacao, Universidade Estadual da Feira de Santana:
Bahia, 2012.
[124] Engelhardt, E.; Brucki, S.M.T.; Cavalcanti, J.L.S.; For-
lenza, O.V.; Laks, J.; Vale, F.A.C.. Tratamento da Doenca
de Alzheimer: Recomendacoes e sugestoes do Depar-
tamento Cientifico da neurologia cognitiva e do envelheci-
mento da academia brasileira de neurologia. Arq Neurop-
siquiatr., 2005, 63(4), 11041112.
[125] Nascimento, E.C.M. Determinacao teorica de propriedades
relevantes para a atividade de inibidor da acetil-
colinesterase. Universidade de Brasilia: Brasilia-DF, 2009.
[126] Greenblatt, H.M.; Kryger, G.; Lewis, T.; Silman, I.; Suss-
man, J.L. Structu re of acetylcholinesterase complexed with
(3)-galanthamine at 2.3A resolution. FEBS Letters, 1999,
463, 321-326.
[127] Sugimoto, H.; Ogura, H.; Arai, Y.; Imura, Y.; Yamanishi,
Y. Research and development of donepezil hydrochloride, a
new type of acetylcholinesterase inhibitor. Jpn. J. Pharma-
col., 2002, 89, 7-20.
[128] Abib Junior, E.; Duarte, L.F.; Pereira, R.; Lemes, A.B.;
Morais, D.C.; Lima, L.G.; Savio, D.; Pengo, S. Estudo de
biodisponibilidade relativa/ bioequivalencia de duas formu-
lacoes de cloridrato de donepezila em voluntarios sadios de
ambos o s sexos. RMB , 2013, 70(1/2), 31-35.
[129] Racchi, M.; Mazzucchelli, M.; Porrello, E.; Lanni, C.; Go-
voni, S. Acetylcholinesterase inhibitors: novel activities of
old molecules. Pharmacological Research, 2004, 50(4),
441-451.
[130] Engelhardt, E.; Bertolucci, P.; Brito-Marques, P. Eficacia
da rivastigmina no desempenho cognitivo de pacientes com
doenca de Alheimer provável leve a moderada: resultados
do braco brasileiro de um estudo aberto multicentrico. Arq.
Neuropsiquiatr., 2003, 61(Suppl), S54-S55.
[131] Bartolucci, C.; Perola, E.; Pilger, C.; Fels, G.; Lamba, D.
Three-dimensional Structures of a Complexo f Gal-
anthamine (Niv alinR) with Acetylcholinesterase From Tor-
pedo californica: Implication for the Design of New Anti-
Alzheimer Drugs. Protein Struct. Funct. Genet., 2001, 42,
182-191.
[132] Hyde, C.; Peters, J.; Bond, M.; Rogers, G.; Hoyle, M.;
Anderson, R.; Jeffreys, M.; Davis, S.; Thokala, P.;
Moxham, T. Evolution of the evidence on the effectiven ess
and cost-effectiveness of acetylcholinesteraseinhibitors and
memantine for Alzheimer’s disease: systematic review and
economic model. Age Ageing, 2013, 42, 14-20.
[133] Forlenza, O.V.; Torres, C.A.; Talib, L.L.; De Paula, V.J.;
Joaquim, H.P.G.; Diniz, B.S.; Gattaz, W.F. Increased plate-
let GSK3B activity in patients with mild cognitive impair-
ment and Alzheimer’s disease. J. Psychiatr. Res., 2011, 45,
220-224.
[134] Epstein, F.H.; Lipton, S.A.; Rosenberg, P.A. Excitatory
Amino Acids as a Final Common Pathway for Neurologic
Disorders. N. Engl. J. Med., 1994, 330(9), 613622.
[135] Bullock, R. Efficacy and Safety of Memantine in Moderate-
to-Severe Alzheimer Disease: The Evidence to Date. Alz-
heimer Dis. Assoc. Disord., 2006, 20(1), 2329.
[136] Miguel-Hidalgo, J.J.; Alvarez, X.A.; Cacabelos, R.; Quack,
G. Neuroprotection by Memantine against Neurodegenera-
tion Induced by B-amyloid(1-40). Brain Res. 2002, 958(1),
210221.
[137] Francis, P.T. Altered Glutamate Neurotransmission and
Behaviour in Dementia: Evidence from Studies of Meman-
tine. Curr. Mol. Pharmacol., 2009, 2(1), 7782.
[138] Fox, C.; Crugel, M.; Maidment, I.; Auestad, B.H.; Coulton,
S.; Treloar, A.; Ballard, C.; Boustani, M.; Katona, C.;
Livingston, G. Efficacy of Memantine for Agitation in Alz-
heimer’s Dementia: A Randomised Double-Blind Placebo
Controlled Trial. PLoS One, 2012, 7(5), e35185.
[139] Gordon, M.L.; Kingsley, P.B.; Goldberg, T.E.; Koppel, J.;
Christen, E.; Keehlisen, L.; Kohn, N.; Davies, P. An Open-
Label Exploratory Study with Memantine: Correlation be-
tween Proton Magnetic Resonance Spectroscopy and Cog-
nition in Patients with Mild to Moderate Alzheimer’s Dis-
ease. Dement. Geriatr. Cogn. Dis. Extra, 2012, 2(1), 312
320.
[140] Kornhuber, J.; Mack-Burkhardt, F.; Riederer, P.; Heben-
streit, G.F.; Reynolds, G. P.; Andrews, H. B.; Beckmann,
H. (3H]MK-801 Binding Sites in Postmortem Brain Re-
gions of Schizophrenic Patients. J. Neural Transm., 1989,
77(2-3), 231236.
[141] Parsons, G. Patch Clamp Studies on the Kinetics and Selec-
tivity of N-Methyl-D-Aspartate Receptor. Neuropharma-
cology, 1993, 32(12), 13371350.
[142] Danysz, W.; Parsons, C.G. The NMDA Receptor Antago-
nist Memantine as a Symptomatological and Neuroprotec-
tive Treatment for Alzheimer’s Disease: Preclinical Evi-
dence. Int. J. Geriatr. Psychiatry, 2003, 18 (SUPPL. 1).
[143] Maidment, I. D.; Fox, C.G.; Boustani, M.; Rodriguez, J.;
Brown, R.C.; Katona, C.L. Efficacy of Memantine on Be-
havioral and Psychological Symptoms Related to Dementia:
A Systematic Meta-Analysis. Ann. Pharmacother., 2008,
42(1), 3238.
[144] Hu, S.; Cui, W.; Mak, S.; Tang, J.; Choi, C.; Pang, Y.; Han,
Y. Bis(propyl)-Cognitin Protects against Glutamate-
Induced Neuro-Excitotoxicity via Concurrent Regulation of
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 17
NO, MAPK/ERK and PI3-K/Akt/GSK3β Pathways. Neuro-
chem. In t., 2013, 62(4), 468477.
[145] Luo, J.; Li, W.; Zhao, Y.; Fu, H.; Ma, D.L.; Tang, J.; Li, C.;
Peoples, R.W.; Li, F.; Wang, Q.; Huang, P.; Xia, J.; Pang,
Y.; Han, Y. Pathologically Activated Neuroprotection via
Uncompetitive Blockade of N-Methyl-D-Aspartate Recep-
tors with Fast off-Rate by Novel Multifunctional Dimer
Bis(propyl)-Cognitin. , 2010, 285(26), 1994719958.
[146] Kamat, P.K.; Rai, S.; Swarnkar, S.; Shukla, R.; Ali, S.;
Najmi, A.K.; Nath, C. Okadaic Acid-Induced Tau Phos-
phorylation in Rat Brain: Role of NMDA Receptor. Neuro-
science, 2013, 238, 97113.
[147] Winblad, B.; Gauthier, S.; Astrom, D.; Stender, K. Meman-
tine Benefits Functional Abilities in Moderate to Severe
Alzheimer’s Disease. J. Nutr. Heal. Aging, 2010, 14(9),
770774.
[148] Dysken, M.W.; Sano, M.; Asthana, S.; Vertrees, J.E.; Pal-
laki, M.; Llorente, M.; Love, S.; Schellenberg, G.D.;
McCarten, J.R.; Malphurs, J.; Prieto, S.; Chen, P.; Loreck,
D.J.; Trapp, G.; Bakshi, R.S.; Mintzer, J.E.; Heidebrink,
J.L.; Vidal-Cardona, A.; Arroyo, L.M.; Cruz, A.R.; Zacha-
riah, S.; Kowall, N.W.; Chopra, M.P.; Craft, S.; Thielke, S.;
Turvey, C.L.; Woodman, C.; Monnell, K.A.; Gordon, K.;
Tomaska, J.; Segal, Y.; Peduzzi, P.N.; Guarino, P.D. Effect
of Vitamin E and Memantine on Functional Decline in Alz-
heimer Disease: The TEAM-AD VA Cooperative Random-
ized Trial. JAMA, 2014, 311(1), 3344.
[149] De Sarno, P.; Bijur, G.N.; Zmijewska, A.A.; Li, X.; Jope,
R.S. In Vivo Regulation of GSK3 Phosphorylation by Cho-
linergic and NMDA Receptors. Neurobiol. Aging, 2006,
27(3), 413422.
[150] Hellweg, R.; Wirth, Y.; Janetzky, W.; Hartmann, S. Effi-
cacy of Memantine in Delaying Clinical Worsening in Alz-
heimer’s Disease (AD): Responder Analyses of Nine Clini-
cal Trials with Patients with Moderate to Severe AD. Int. J.
Geriatr. Psychiatry, 2012, 27(6), 651656.
[151] Hemmati, F.; Dargahi, L.; Nasoohi, S.; Omidbakhsh, R.;
Mohamed, Z.; Chik, Z.; Naidu, M.; Ahmadiani, A. Neuror-
estorative Effect of FTY720 in a Rat Model of Alzheimer’s
Disease: Comparison with Memantine. Behav. Brain Res.,
2013, 252, 415421.
[152] Forlenza, O.V. Tratamento farmacologico da doenca de
Alzheimer. Rev. Psiq. Clin., 2005, 32(3), 137-148.
[153] Sinha, S.; Liebenburg, I. Cellular mechanisms of β-amyloid
production and secretion. Proc. Natl. Acad. Sci., 1999, 96,
1104-11053.
[154] Zhu, Y.; Xiao, K.; Ma, L.; Xiong, B.; Fu, Y.; Yu, H.; Wang,
W.; Wang, X.; Hu, D.; Peng, H.; Li, J.; Gong, Q.; Chai, Q.;
Tang, X.; Zhang, H.; Li, J.; Shen, J. Design, synthesis and
biological evaluation of novel dual inhibitors of acetlcho-
linesterase and β-secrease. Bioorg. Med. Chem., 2009, 17,
1600-1613.
[155] Zhang, C. Natural Compounds That Modulate BACE1-
processing of Amyloid-beta Precursor Protein in Alz-
heimer’s Disease. Discov. Med., 2012, 14(76), 189-197.
[156] Gosh, A.K.; Brindisi, M.; Tang, J. Developing β-secrease
inhibitors for treatment of Alzheimer's disease. J. Neuro-
chem., 2012, 120(Suppl 1), 71-83.
[157] Gosh, A.K.; Kumaragurubaran, N.; Hong, L.; Kulkarni, S.;
Xu, X.; Miller, H.B.; Reddy, D.S.; Weerasena, V.; Turner,
R.; Chang, W.; Koelsch, G.; Tang, J. Bioorg. Med. Chem.
Lett., 2008, 18, 1031-1036.
[158] Steele, T.G.; Hills, I.D.; Nomland, A.A.; Leon, P.; Allison,
T.; Mcgaughey, G.; Colussi, D.; Tugusheva, K.; Hauga-
book, S.J.; Espeseth, A.S.; Zuck, P.; Graham, S.L.; Stachel,
S.J. Identification of small molecule β-secrease inhibitor
that binds without catalytic aspartate engagement. Bioorg.
Med. Chem., 2009, 19, 17-20.
[159] Xiao, K.; Li, X.; Li, J.; Ma, L.; Hu, B.; Yu, H.; Fu, Y.;
Wang, R.; Ma, Z.; Qiu, B.; Li, Jia.; Hu, D.; Wang, X.; Shen,
J. Design, synthesis, and evaluation of Leu*Ala hy-
droxyethylene-based non-peptide β-secrease (BACE) in-
hibitors. Bioorg. Med. Chem., 2006, 14, 4535-4551.
[160] Siemers, E.; Dean, R.A.; Satterwhite, J.; Farlow, M.R.;
Skinner, M.; Ness, D.; May, P. C. Safety, tolerability and
changes in plasma, and cerebrospinal fluid amyloid beta
concentrations after administration of a functional gamma-
secretase inhibitor in healthy volunteers. Neurobiol. Aging,
2004, 25(S2), S569-S570.
[161] Hu, S.; Begum, A.N.; Jones, M.R.; Oh, M.S.; Beech, W.K.;
Beech, B.H.; Yang, F.; Chen, P.; Ubeda, O.J.; Kim, P.C.;
Davies, P.; Ma, Q.; Cole, G.M.; Frautschy, S.A. GSK3 In-
hibitors Show Benefits in an Alzheimer’s Disease (AD)
Model of Neurodegeneration but Adverse Effects in Control
Animals. Neurobiol. Dis., 2009, 33(2), 193206.
[162] Wood-Kaczmar, A.; Kraus, M.; Ishiguro, K.; Philpott, K.L.;
Gordon-Weeks, P. R. An Alternatively Spliced Form of
Glycogen Synthase Kinase-3β Is Targeted to Growing Neu-
rites and Growth Cones. Mol. C ell. Neurosci., 2009, 42(3),
184194.
[163] Castano, Z.; Gordon-Weeks, P.R.; Kypta, R.M. The Neu-
ron-Specific Isoform of Glycogen Synthase Kinase-3beta Is
Required for Axon Growth. J.Neurochem., 2010, 113(1),
117130.
[164] Cai, Z.; Zhao, Y.; Zhao, B. Roles of Glycogen Synthase
Kinase 3 in Alzheimer’s Disease. Curr. Alzheimer Res.,
2012, 9(7), 864879.
[165] Grimes, C.A; Jope, R.S. The Multifaceted Roles of Glyco-
gen Synthase Kinase 3beta in Cellular Signaling. Prog.
Neurobiol., 2001, 65(4), 391426.
[166] Chen, Y.Y.; Chen, G.; Fan, Z.; Luo, J.; Ke, Z.J. GSK3β and
Endoplasmic Reticulum Stress Mediate Rotenone-Induced
Death of SK-N-MC Neuroblastoma Cells. Biochem. Phar-
macol., 2008, 76(1), 128138.
[167] Hongo, H.; Kihara, T.; Kume, T.; Izumi, Y.; Niidome, T.;
Sugimoto, H.; Akaike, A. Glycogen Synthase Kinase-3β
Activation Mediates Rotenone-Induced Cytotoxicity with
the Involvement of Microtubule Destabilization. Biochem.
Biophys. Res. Commun., 2012, 426(1), 9499.
[168] Gendron, T.F.; Petrucelli, L. The Role of Tau in
Neurodegeneration. Mol. Neurodegener., 2009, 4, 13.
[169] Fukunaga, K.; Uehara, F.; Aritomo, K.; Shoda, A; Hiki, S.;
Okuyama, M.; Usui, Y.; Watanabe, K.; Yamakoshi, K.;
Kohara, T.; Hanano, T.; Tanaka, H.; Tsuchiya, S.; Sunada,
S.; Saito, K.; Eguchi, J.; Yuki, S.; Asano, S.; Tanaka, S.;
Mori, A; Yamagami, K.; Baba, H.; Horikawa, T.; Fujimura,
M. 2-(2-Phenylmorpholin-4-Yl)pyrimidin-4(3H)-Ones; a
New Class of Potent, Selective and Orally Active Glycogen
Synthase Kinase-3beta Inhibitors. Bioorg Med Chem Lett.,
2013, 23(24), 69336937.
[170] Martinez, A.; Alonso, M.; Castro, A.; Perez, C.; Moreno,
F.J. First Non-ATP Competitive Glycogen Synthase Kinase
3 Beta (GSK-3beta) Inhibitors: Thiadiazolidinones (TDZD)
as Potential Drugs for the Treatment of Alzheimer’s Dis-
ease. J. Med. Chem., 2002, 45(6), 12921299.
[171] Hers, I.; Tavare, J.M.; Denton, R.M. The Protein Kinase C
Inhibitors Bisindolylmaleimide I (GF 109203x) and IX (Ro
31-8220) Are Potent Inhibitors of Glycogen Synthase
Kinase-3 Activity. FEBS Lett., 1999, 460(3), 433436.
[172] Smith, D.G.; Buffet, M.; Fenwick, A.E.; Haigh, D.; Ife,
R.J.; Saunders, M.; Slingsby, B.P.; Stacey, R.; Ward, R.W.
3-Anilino-4-Arylmaleimides: Potent and Selective Inhibi-
tors of Glycogen Synthase Kinase-3 (GSK-3). Bioorg. Med.
Chem. Lett., 2001, 11(5), 635639.
[173] Leost, M.; Schultz, C.; Link, A.; Wu, Y.Z.; Biernat, J.;
Mandelkow, E.M.; Bibb, J.A; Snyder, G.L.; Greengard, P.;
18 Current Medicinal Chemistry, 2017, Vol. 24, No. 00 dos Santos Picanço et al.,
Zaharevitz, D.W.; Gussio, R.; Senderowicz, A.M.;
Sausville, E.A; Kunick, C.; Meijer, L. Paullones Are Potent
Inhibitors of Glycogen Synthase Kinase-3β and Cyclin-
Dependent Kinase 5/p25. Eur. J. Biochem., 2000, 267(19),
59835994.
[174] Leclerc, S.; Garnier, M.; Hoessel, R.; Marko, D.; Bibb,
J.A.; Snyder, G.L.; Greengard, P.; Biernat, J.; Wu, Y.Z.;
Mandelkow, E.M.; Eisenbrand, G.; Meijer, L. Indirubins
Inhibit Glycogen Synthase Kinase-3beta and CDK5/P25,
Two Protein Kinases Involved in Abnormal Tau Phos-
phorylation in Alzheimer’s Disease. A Property Common to
Most Cyclin-Dependent Kinase Inhibitors? J. Biol. Chem.,
2001, 276(1), 251260.
[175] Meijer, L.; Thunnissen, A.M.; Wh ite, A.; Garnier, M.; Nik-
olic, M.; Tsai, L.-H.; Walter, J.; Cleverley, K.; Salinas, P.;
Wu, Y.Z.; Biernat, J.; Mandelkow, E.-M.; Kim, S.-H.; Pet-
tit, G. Inhibition of Cyclin-Dependent Kinases, GSK-3β and
CK1 by Hymenialdisine, a Marine Sponge Constituent.
Chem. Biol., 2000, 7(1), 5163.
[176] Wang, M.; Gao, M.; Miller, K.D.; Sledge, G.W.; Hutchins,
G.D.; Zheng, Q.H. The First Synthesis of [11C]SB-216763,
a New Potential PET Agent for Imaging of Glycogen Syn-
thase Kinase-3 (GSK-3). Bioorg. Med. Chem. Lett., 2011,
21(1), 245249.
[177] Thotala, D.K.; Hallahan, D.E.; Yazlovitskaya, E.M. Inhibi-
tion of Glycogen Synthase Kinase 3β Attenuates Neurocog-
nitive Dysfunction Resulting from Cranial Irradiation. Can-
cer Res., 2008, 68(14), 58595868.
[178] Ryves, W.J.; Harwood, A.J. Lithium Inhibits Glycogen
Synthase Kinase-3 by Competition for Magnesium. Bio-
chem. Biophys. Res. Commun., 2001, 280(3), 720725.
[179] Aghdam, S.Y.; Barger, S.W. Glycogen Synthase Kinase-3
in Neurodegeneration and Neuroprotection: Lessons from
Lithium. Curr. Alzheimer Res., 2007, 4(1), 2131.
[180] Martinez, A. Preclinical Eficacy on GSK-3 Inhibitors: To-
wards a Future Generation of Powerful Drugs. Med. Res.
Rev., 2008, 28(5), 773796.
[181] Noble, W.; Planel, E.; Zehr, C.; Olm, V.; Meyerson, J.;
Suleman, F.; Gaynor, K.; Wang, L.; LaFrancois, J.; Fein-
stein, B.; Burn s, M.; Krishnamurthy, P.; Wen, Y.; Bhat, R.;
Lewis, J.; Dickson, D.; Duff, K. Inhibition of Glycogen
Synthase Kinase-3 by Lithium Correlates with Reduced
Tauopathy and Degeneration in Vivo. Proc. Natl. Acad. Sci.
U. S. A., 2005, 102(19), 69906995.
[182] Tariot, P.N.; Aisen, P.S. Can Lithium or Valproate Untie
Tangles in Alzheimer’s Disease? Journal of Clinical Psy-
chiatry. 2009, 70(6), 919921.
[183] Fleisher, A.S.; Truran, D.; Mai, J.T.; Langbaum, J.B.S.;
Aisen, P.S.; Cummings, J.L.; Jack, C.R.; Weiner, M.W.;
Thomas, R.G.; Schneider, L.S.; Tariot, P. N. Chronic Di-
valproex Sodium Use and Brain Atrophy in Alzheimer Dis-
ease. Neurology, 2011, 77, 12631271.
[184] Avrahami, L.; Farfara, D.; Shaham-Kol, M.; Vassar, R.;
Frenkel, D.; Eldar-Finkelman, H. Inhibition of Glycogen
Synthase Kinase-3 Ameliorates β- Amyloid P athology and
Restores Lysosomal Acidification and Mammalian Target
of Rapamycin Activity in the Alzheimer Disease Mouse
Model: In Vivo and in Vitro Studies. J. Biol. Chem., 2013,
288(2), 12951306.
[185] Berg, S.; Bergh, M.; Hellberg, S.; Hogdin, K.; Lo-
Alfredsson, Y.; Soderman, P.; von Berg, S.; Weigelt, T.;
Ormo, M.; Xue, Y.; Tucker, J.; Neelissen, J.; Jerning, E.;
Nilsson, Y.; Bhat, R. Discovery of Novel Potent and Highly
Selective Glycogen Synthase Kinase-3beta (GSK3beta) In-
hibitors for Alzheimer’s Disease: Design, Synthesis, and
Characterization of Pyrazines. J. M ed. Chem., 2012, 55(21),
91079119.
[186] Coffman, K.; Brodney, M.; Cook, J.; Lanyon, L.; Pandit, J.;
Sakya, S.; Schachter, J.; Tseng-Lovering, E.; Wessel, M. 6-
Amino-4-(Pyrimid in-4-Yl)pyridones: Novel Glycogen Syn-
thase Kinase-3β Inhibitors. Bioorg. Med. Chem. Lett., 2011,
21(5), 14291433.
[187] Zhang, M.R.; Maeda, J.; Ogawa, M.; Noguchi, J.; Ito, T.;
Yoshida, Y.; Okauchi, T.; Obayashi, S.; Suhara, T.; Suzuki,
K. Development of a New Radioligand, N -(5-Fluoro-2-
Phenoxyphenyl)- N -(2-[ 18 F]fluoroethyl-5-
Methoxybenzyl)acetamide, for PET Imaging of Peripheral
Benzodiazepine Receptor in Primate Brain. J. Med. Chem.,
2004, 47(9), 22282235.
[188] Saitoh, M.; Kunitomo, J.; Kimura, E.; Iwashita, H.; Uno,
Y.; Onishi, T.; Uchiyama, N.; Kawamoto, T.; Tanaka, T.;
Mol, C.D.; Dougan, D.R.; Textor, G. P.; Snell, G.P.; Taki-
zawa, M.; Itoh, F.; Ko ri, M. 2-{3-[4-(Alkylsu lfinyl)phenyl]-
1-Benzofuran-5-yl}-5-Methyl-1,3,4-Oxadiazole Derivatives
as Novel Inhibitors of Glycogen Synthase Kinase-3β with
Good Brain Permeability. J. Med. Chem., 2009, 52(20),
62706286.
[189] Onishi, T.; Iwashita, H.; Uno, Y.; Kunitomo, J.; Saitoh, M.;
Kimura, E.; Fujita, H.; Uchiyama, N.; Kori, M.; Takizawa,
M. A Novel Glycogen Synthase Kinase-3 Inhibitor 2-
Methyl-5-(3-{4-[(S)-Methylsulfinyl]phenyl}-1-Benzofuran-
5-yl)-1,3,4-Oxadiazole Decreases Tau Phosphorylation and
Ameliorates Cognitive Deficits in a Transgenic Model of
Alzheimer’s Disease. J. Neuroch em., 2011, 119(6), 1330
1340.
[190] Kumata, K.; Yui, J.; Xie, L.; Zhang, Y.; Nengaki, N.; Fuji-
naga, M.; Yamasaki, T.; Shimoda, Y.; Zhang, M.-R. Radio-
synthesis and Preliminary PET Evaluation of Glycogen
Synthase Kinase 3β (GSK-3β) Inhibitors Containing
[11C]methylsulfanyl, [11C]methylsulfinyl or
[11C]methylsulfonyl Groups. Bioorg. Med. Chem. Lett.,
2015, 25(16), 32303233.
[191] Lu, J.; Maezawa, I.; Weerasekara, S.; Erenler, R.; Nguyen,
T.D.T.; Nguyen, J.; Swisher, L.Z.; Li, J.; Jin, L. W.; Ran-
jan, A.; Srivastava, S. K.; Hua, D. H. Syntheses, Neural
Protective Activities, and Inhibition of Glycogen Synthase
Kinase-3β of Substituted Quinolines. Bioorg. Med. Chem.
Lett., 2014, 24(15), 33923397.
[192] Ma, S.L.; Pastorino, L .; Zhou, X.Z.; Lu, K.P. Prolyl
Isomerase Pin1 Promotes Amyloid Precursor Pro tein (APP)
Turnover by Inhibiting Glycogen Synthase Kinase-3β
(GSK3β) Activity: Novel Mechanism for Pin1 to Protect
against Alzh eimer Disease. J. Biol. Chem., 2012, 287(10),
69696973.
[193] Noh, M.Y.; Chun, K.; Kang, B. Y.; Kim, H.; Park, J.-S.;
Lee, H.-C.; Kim, Y.-H.; Ku, S.; Kim, S. H. Newly Devel-
oped Glycogen Synthase Kinase-3 (GSK-3) Inhibitors Pro-
tect Neuronal Cells Death in Amyloid-Beta Induced Cell
Model and in a Transgenic Mouse Model of Alzheimer’s
Disease. Biochem. Biophys. Res. Commun., 2013, 435(2),
274281.
[194] Uehara, F.; Shoda, A.; Aritomo, K.; Fukunaga, K.; Wata-
nabe, K.; Ando, R.; Shinoda, M.; Ueno, H.; Kubodera, H.;
Sunada, S.; Saito, K. I.; Kaji, T.; Asano, S.; Eguchi, J. I.;
Yuki, S.; Tanaka, S.; Yoneyama, Y.; Niwa, T. 6-(4-
Pyridyl)pyrimidin-4(3H)-Ones as CNS Penetrant Glycogen
Synthase Kinase-3β Inhibitors. Bioorg. Med. Chem. Lett.,
2013, 23(24), 69286932.
[195] Giacobini, E.; Gold, G. Alzheimer disease therapy
moving from amyloid β to tau. Nat. Rev. Neurol., 2013,
9(12), 677-686.
[196] Da Rocha, M.D.; Viegas, F.P.D.; Campos, H.C.; Nicastro,
P.C.; Fossaluzza, P.C.; Fraga, C.A.M.; Barreiro, E.J; Viegas
Jr, C. The role of natural products in the discovery of new
drug candidates for the treatment of neurodegenerative dis-
Alzheimer's Disease: A Review from the Pathophysiology to Diagnosis Current Medicinal Chemistry, 2017, Vol. 24, No. 00 19
orders II: Alzheimer’s disease. CNS Neurol. Disord. Drug
Targets, 2011, 10(2), 251270.
[197] Moser, D.A. Mal de Alzheimer na primeira fase: contribui-
coes da psicolinguistica. Tese, Universidade Federal de
Santa Catarina: Florianopolis, 2011.
[198] Dias, K.S.T.; De Paula, C.T.; Riquiel, M.M.; Lago, S.T.;
Costa, K.C.M.; Vaz, S.M.; Machado, R.P.; Lima, L.M.S.;
Viegas Junior, C. Recent Applications of the Multi-Target
Directed Ligands Approach for the Treatment of Alz-
heimer’s Disease. Rev. Virtual Quim., 2015, 7(2), 609-648.
[199] Lim, J. W.; Kim, H.; Kim, K. H. Nuclear factor-kappa B
regulates cyclooxygenase-2 expression and cell prolifera-
tion in human gastric cancer cells. Lab. Invest. 2001, 81,
349360.
[200] Aisen, P.S.; Schafer, K.A.; Grundmn, M.; Pfeiffer, E.;
Sano, M.; Davis, K.L.; Farlow, M.R.; Jin, S.; Thomas,
R.G.; Thal, L.J. Effects of rofecoxib or naproxen vs placebo
on Alzheimer disease progression: a randomized controlled
Trial. JAMA, 2003, 289(21), 2819-2826.
[201] Gertsch, J.; Anavi-Goffer, S. Metilhonokiol attenuates neu-
roinflammation: a role for cannabinoid receptors? J. Neu-
roinflammation, 2012, 9, 135-140.
... It is one of the main contributors to dementia cases worldwide. About 5.3 million human being in the US alone have AD, of which 5.1 million are 65 or older and 200,000 have the disease at a younger age [9]. Li N.M et al. ...
Chapter
Extracellular amyloid-(A) plaques and neurofibrillary in the intracellular environment are defining characteristics of Alzheimer's disease (AD), neuronal death, and synaptic loss, all of which contribute to progressive cognitive impairment. Age is the most significant risk factor for AD, with AD prevalence exponentially increasing after age 65. As the average lifespan in emerging countries rises, it is anticipated that the total prevalence of AD will double during the next 20 years. Effective drug delivery to the target tissue, constant therapeutic drug concentrations, a lowering in dosing quantity and frequency, and increased patient compliance are all qualities of ideal drug delivery systems which depend on Pharmacokinetics and Pharmacodynamic of Various novel formulations Targeting AD. Currently different formulations of Donepezil Hydrochloride; Memantine Hydrochloride are available in market such tablets orally disintegrant, Capsule extended release and lots of attempts are going on in term of different delivery system like liposomal drug delivery, nose to brain delivery system, nanoformulations having different nanocarriers for the treatment of AD. Fate of all the novel formulations depends on the Pharmacokinetics and Pharmacodynamic behavior of the drug substances. Pharmacokinetics (PK) studies the disposition of drug molecules in the body, including their concentration patterns and AUC last, Tmax, Cmax, and AUC inf values Pharmacodynamic (PD) assessment looks at how a drug affects the body. includes variations in Emax, EC50, in an Emax-model parameter. A drug must cross the blood brain barriers and be absorbed by the specific tissues in order for it to be effective in treating an Alzheimer patient (as measured by PK studies). To successfully alter the target protein activity in the body (as determined by PD studies), this is required. This chapter will focus on the various novel formulations Targeting AD and its Pharmacokinetics and Pharmacodynamics behavior.
Article
Self‐assembled aggregation of peptides and proteins into regular amyloid fibrils is associated with several neurodegenerative diseases. In case of Alzheimer's disease proteolytic cleavage products of the amyloid precursor protein form pathological amyloid‐beta fibrils in a nucleation and propagation phase. The molecular details and thermodynamic driving forces of amyloid formation are not well understood, but are of high relevance for potential pharmacological interference. We used atomistic binding free energy simulations to calculate the free energy of protofilament propagation by an additional Aβ 9–40 peptide binding to the protofilament tip. It requires sampling of relevant conformational transitions which is challenging since the monomeric Aβ 9–40 peptide is intrinsically disordered. However, the convergence of umbrella simulations can be enhanced by applying additional restraining potentials on the axial, orientational and conformational degrees of freedom. The improved convergence leads to a much closer agreement with experimental binding free energy data compared to unrestrained umbrella sampling. Moreover, the restraining approach results in a separation of contributions to the total binding free energy. The calculated contributions indicate that the free energy change associated with the restriction of conformational freedom upon propagation makes a large opposing contribution of higher magnitude than the total binding free energy. Finally, optimization of the approach leads to further significant reduction of the computational demand which is crucial for systematic studies on mutations, denaturants and inhibitors in the fibril propagation step.
Article
Background: Alzheimer's disease (AD) is characterized by disrupted proteostasis and macroautophagy (hereafter "autophagy"). The pharmacological agent suramin has known autophagy modulation properties with potential efficacy in mitigating AD neuronal pathology. Objective: In the present work, we investigate the impact of forebrain neuron exposure to suramin on the Akt/mTOR signaling pathway, a major regulator of autophagy, in comparison with rapamycin and chloroquine. We further investigate the effect of suramin on several AD-related biomarkers in sporadic AD (sAD)-derived forebrain neurons. Methods: Neurons differentiated from ReNcell neural progenitors were used to assess the impact of suramin on the Akt/mTOR signaling pathway relative to the autophagy inducer rapamycin and autophagy inhibitor chloroquine. Mature forebrain neurons were differentiated from induced pluripotent stem cells (iPSCs) sourced from a late-onset sAD patient and treated with 100μM suramin for 72 h, followed by assessments for amyloid-β, phosphorylated tau, oxidative/nitrosative stress, and synaptic puncta density. Results: Suramin treatment of sAD-derived neurons partially ameliorated the increased p-Tau(S199)/Tau ratio, and fully remediated the increased glutathione to oxidized nitric oxide ratio, observed in untreated sAD-derived neurons relative to healthy controls. These positive results may be due in part to the distinct increases in Akt/mTOR pathway mediator p-p70S6K noted with suramin treatment of both ReNcell-derived and iPSC-derived neurons. Longer term neuronal markers, such as synaptic puncta density, were unaffected by suramin treatment. Conclusions: These findings provide initial evidence supporting the potential of suramin to reduce the degree of dysregulation in sAD-derived forebrain neurons in part via the modulation of autophagy.
Conference Paper
Alzheimer’s Disease (AD) is a typical neuro-degenerative illness. Numerous studies have discovered that the development of AD could produce obvious changes in brain structure and cerebrospinal fluid (CSF). With obtained brain features, our study proposed an approach of Deep Residual Network (DRN) with brain imaging and CSF features for AD classification. To accommodate the input of one-dimensional data, we opted to substitute the convolutional layer in our model with the dense layer. Specifically, the accuracy rates for the AD/NC, AD/MCI, and MCI/NC groups stood at 92.50%, 80.0%, and 67.50% respectively. Moreover, the three-class classification achieved an accuracy of 63.33%. For performance evaluation, our model was superior to those of other machine learning (ML) models. The proposed model demonstrated robust capability in revealing the potential biomarkers for AD classification and early predication.
Chapter
The book summarizes the role of multiple enzyme targets and strategies to design and develop novel drug candidates for Alzheimer's disease (AD). Insights from researchers across the globe from diverse fields are presented in a thematic volume. The chapters highlight current information scientists have unraveled about the origin, pathogenesis and prevention of AD. The contributions consider both established and emerging drug targets viz. Tau proteins, TREM, and microglia. Topics covered in the book include multi-target anti-Alzheimer's agents, epigenetic modifications, and the role of specific proteins like TMP21 and Tau in AD. A section dedicated to pharmacological treatments discusses the significance of tubulin-modifying enzymes, memantine, and glutamate antagonists. Enzymatic targets for drug discovery are thoroughly examined, focusing on cholinesterase, secretases, and other enzymes. Additionally, the book explores innovative nano-carrier-based drug delivery methods, emphasizing the crucial role of nanotechnology in effective Alzheimer's treatment. The book aims to inform students and researchers in the field of neuroscience, medicine and pharmacology about current research and biochemical nuances of AD pathogenesis and enzymatic drug targeting strategies.
Chapter
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by multifactorial processes related to different biological targets. Acetylcholinesterase (AChE) inhibitors have been used in AD treatment, but other therapeutic alternatives have prospected, such as Glycogen Synthase Kinase-3β (GSK-3β) inhibitors. Considering the limitations of one-target drugs, the use of multi-target directed drugs rises as a promising strategy. Design new AChE-GSK-3β dual inhibitors as propitious multi-target drug candidates for AD treatment. Thirteen natural products were selected, molecular docking, molecular dynamics, pharmacophore modeling, ADME/Tox predictions and proposals development was performed. Given the results obtained and considering structural subunits of AChE and GSK-3β inhibitors, structural modifications were done to design new hybrids proposals. Hydrophobic and hydrogen bond interactions between compounds and important residues of AChE and GSK-3β were observed, besides, three pharmacophoric features were founded: one aromatic ring and two hydrogen bond acceptors. Compounds also demonstrated favorable ADME/Tox predictions. Subsequently, these results afforded the use of physostigmine and luteolin as prototypes for the design of nine hybrid proposals: five AChE inhibitors and four AChE-GSK-3β dual-inhibitors. Their bioactivity prediction showed that all proposals should exhibit anti-Alzheimer related activity; ADME/Tox predictions showed best results for Proposals 1, 2 and 5; and Proposals 6, 7, 8 and 9 presented higher affinity for both AChE and GSK-3β enzymes. Proposals 1 and 2 were considered potential AChE inhibitors, and Proposals 6 and 8 were elected as potential AChE-GSK-3β dual inhibitors for therapeutics in AD.
Patent
Full-text available
This invention corresponds to an in vitro method that employs an electrophysiology technique; in particular, an embodiment of the patch-clamp technique in its perforated type, which will allow us evaluating the neurotoxic capacity of protein structures associated to the generation of neurodegenerative diseases. It addition, it allows evaluating potential pharmacologic capacities of candidate molecules (drugs) in order to prevent, treat, or cure the said diseases. The invention involves the use of peptides that cause neurodegenerative diseases in order to form a spontaneous perforated recording that only occurs with peptides in the patch clamp glass pipette.
Article
Full-text available
Early signs of dementia There is currently no cure for Alzheimer's disease. One of the reasons could be that interventions start too late, when there is already irreversible damage to the brain. Developing a biomarker that would help to effectively start therapy at very early stages of the disease is thus of high interest. Kunz et al. studied neural correlates of spatial navigation in the entorhinal cortex in control study participants and individuals at risk of developing Alzheimer's. The at-risk group showed a different brain signal many decades before the onset of the disease, and they navigated differently in a virtual environment. Science , this issue p. 430
Article
A Doença de Alzheimer (DA) é uma patologia neurodegenerativa caracterizada por prejuízo das funções cognitivas e mudanças na personalidade, responsável por cerca de 50-60% dos casos de demência diagnosticados em pessoas com mais de 60 anos. O depósito cerebral de um peptídeo insolúvel, chamado peptídeo β-amilóide, e a presença de emaranhados neurofibrilares são considerados os marcadores neuropatológicos da doença. Somente cinco fármacos estão aprovados pelo FDA para o tratamento da DA, sendo que nenhum pode reverter o processo de neurodegeneração provocado pela doença. Atualmente o tratamento consiste apenas em aliviar temporariamente os sintomas, ou seja, não há cura para a DA. Portanto, em busca de substâncias potencialmente úteis para o tratamento da DA, os produtos naturais têm se destacado por apresentarem um papel importante na patologia da doença, não agindo somente na causa, mas também em seus mecanismos específicos.
Article
Simplified explanation of exam procedures such as assessment of level of consciousness, language acuity, memory, and attention Pocket card summarizes the main points to remember in assessing mental status in limited patient-contact time DSM-IV diagnostic criteria are covered to ease filling out insurance forms Findings about cognition and testing a patient's mental function Covers the refinement of testing procedures and the psychological test battery to speed up the exam Presents age-related norms, important when assessing the elderly for early dementia Reviews ways to use the exam as a screening procedure, particularly when diagnosing dementia and differentiating between organic and functional disorders
Article
For over fifty years lithium has been a fundamental component of therapy for patients with bipolar disorders. Lithium has been considered recently for its potential to alleviate neuronal loss and other neurodegeneration processes. For instance, lithium reduces the severity of some behavioral complications of Alzheimer's disease (AD). And there are growing indications that lithium may be of benefit to the underlying pathology of AD, as well as an array of other common CNS disorders, including stroke, Parkinson's disease, and Huntington's disease. Despite these demonstrated and prospective therapeutic benefits, lithium's mechanism of action remains elusive, and opinions differ regarding the most relevant molecular targets. Lithium inhibits several enzymes; significant among these are inositol monophosphatase (IMPase), glycogen synthase kinase-3 (GSK-3), and the proteasome. Most recent publications discussing the medical application of lithium have converged on GSK-3, so this article reviews data and discussions regarding the roles and interactions of GSK-3 with other proteins and its proposed role in the pathogenesis of Alzheimer's disease.
Article
Evidence from basic molecular biology has noted a critical role of GSK-3 in Alzheimer's disease (AD) pathogenesis such as beta-amyloid (A beta) production and accumulation, the formation of neurofibrillary tangle (NFT), and neuronal degeneration. A beta generation and deposition represents a key feature and is generated from APP by the sequential actions of two proteolytic enzymes: beta-secretase and gamma-secretase. GSK-3 could play a critical role in A beta production via enhancing beta-secretase activity. GSK-3 not only modulates APP processing in the process of A beta generation, but regulates A beta production by interfering with APP cleavage at the gamma-secretase complex step since the APP and PS1 (a component of gamma-secretase complex) are substrates of GSK-3 as well. GSK-3 may downregulate alpha-secretase through inhibiting PKC and ADAMs activity which are the substrates of GSK-3 contributing to A beta production. Meanwhile, A beta accumulation can induce GSK-3 activation through A beta-mediated neuroinflammation and oxidative stress. Considering that active GSK-3 and some common GSK-3-shared factors induce the hyperphosphorylation of tau and neurofibrillary lesions, GSK-3 is a possible linking between amyloid plaques and NFT pathology. Additionally, GSK-3 could disrupt acetylcholine activity, and accelerate axon degeneration and failures in axonal transport, and lead to cognitive impairment in AD. Preclinical and clinical studies have supported that GSK-3 beta inhibitors could be useful in the treatment of AD. Consequently, an effective measure to inhibit GSK-3 activity may be a very attractive drug target in AD.