ArticlePublisher preview availableLiterature Review

Multi-Target Drug Candidates for Multifactorial Alzheimer’s Disease: AChE and NMDAR as Molecular Targets

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Alzheimer’s disease (AD) is one of the most common forms of dementia among elder people, which is a progressive neurodegenerative disease that results from a chronic loss of cognitive activities. It has been observed that AD is multifactorial, hence diverse pharmacological targets that could be followed for the treatment of AD. The Food and Drug Administration has approved two types of medications for AD treatment such as cholinesterase inhibitors (ChEIs) and N-methyl-D-aspartic acid receptor (NMDAR) antagonists. Rivastigmine, donepezil, and galantamine are the ChEIs that have been approved to treat AD. On the other hand, memantine is the only non-competitive NMDAR antagonist approved in AD treatment. As compared with placebo, it has been revealed through clinical studies that many single-target therapies are unsuccessful to treat multifactorial Alzheimer’s symptoms or disease progression. Therefore, due to the complex nature of AD pathophysiology, diverse pharmacological targets can be hunted. In this article, based on the entwined link of acetylcholinesterase (AChE) and NMDAR, we represent several multifunctional compounds in the rational design of new potential AD medications. This review focus on the significance of privileged scaffolds in the generation of the multi-target lead compound for treating AD, investigating the idea and challenges of multi-target drug design. Furthermore, the most auspicious elementary units for designing as well as synthesizing hybrid drugs are demonstrated as pharmacological probes in the rational design of new potential AD therapeutics.
This content is subject to copyright. Terms and conditions apply.
Multi-Target Drug Candidates for Multifactorial Alzheimers Disease:
AChE and NMDAR as Molecular Targets
Md. Sahab Uddin
1,2
&Abdullah Al Mamun
1,2
&Md. Tanvir Kabir
3
&Ghulam Md Ashraf
4,5
&May N. Bin-Jumah
6
&
Mohamed M. Abdel-Daim
7,8
Received: 29 July 2020 /Accepted: 2 September 2020
#Springer Science+Business Media, LLC, part of Springer Nature 2020
Abstract
Alzheimers disease (AD) is one of the most common forms of dementia among elder people, which is a progressive neurode-
generative disease that results from a chronic loss of cognitive activities. It has been observed that AD is multifactorial, hence
diverse pharmacological targets thatcould be followed for the treatment of AD. The Food and Drug Administration has approved
two types of medications for AD treatment such as cholinesterase inhibitors (ChEIs) and N-methyl-D-aspartic acid receptor
(NMDAR) antagonists. Rivastigmine, donepezil, and galantamine are the ChEIs that have been approved to treat AD. On the
other hand, memantine is the only non-competitive NMDAR antagonist approved in AD treatment. As compared with placebo, it
has been revealed through clinical studies that many single-target therapies are unsuccessful to treat multifactorial Alzheimers
symptoms or disease progression. Therefore, due tothe complex nature of AD pathophysiology,diverse pharmacological targets
can be hunted. In this article, based on the entwined link of acetylcholinesterase (AChE) and NMDAR, we represent several
multifunctional compounds in the rational design of new potential AD medications. This review focus on the significance of
privileged scaffolds in the generation of the multi-target lead compound for treating AD, investigating the idea and challenges of
multi-target drug design. Furthermore, the most auspicious elementary units for designing as well as synthesizing hybrid drugs
are demonstrated as pharmacological probes in the rational design of new potential AD therapeutics.
Keywords Alzheimers disease .Multi-target drugs .Multi-target-directed ligands .AChE .NMDAR
Introduction
Alzheimers disease (AD) is the utmost recurrent cause of
dementia that causes a severe loss of cognitive functions [1,
2]. This multifactorial neurodegenerative disorder is charac-
terized by nerve cell death, intracellular neurofibrillary tangles
(NFTs), and extracellular amyloid plaques [36]. It is well
known that these plaques are made of amyloid beta (Aβ),
which is a cleavage product of amyloid precursor protein
(APP) [7]. Furthermore, Aβmonomers gradually aggregate
into oligomers, fibrils, and insoluble amyloid plaques [8,9].
NFTs are aggregates of the hyperphosphorylated tau protein.
The stabilization of microtubules is promoted by tau under
normal conditions [10]. However, hyperphosphorylated tau
accumulates as paired helical filaments that subsequently
leads to NFTs [11,12]. Neuronal and synaptic functions are
dysregulated due to Aβaccumulation, which further makes
the intracellular conditions for NFTs formation eventu-
ally leading to neuronal loss that lead to the disturbance
in the functions of neurotransmitters [1315].
In the basal forebrain, cholinergic neuron loss is assumed
to mediate cholinergic impairment, which eventually leads to
short-term memory loss in AD [16,17]. Thus, a deterioration
in the cholinergic markers and acetylcholine (ACh) are well-
reported in the brains of individuals with AD [1820]. It was
observed that neuronal cell survival was compromised due to
*Md. Sahab Uddin
msu-neuropharma@hotmail.com; msu_neuropharma@hotmail.com
1
Department of Pharmacy, Southeast University, Dhaka, Bangladesh
2
Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
3
Department of Pharmacy, Brac University, Dhaka, Bangladesh
4
King Fahd Medical Research Center, King Abdulaziz University,
Jeddah, Saudi Arabia
5
Department of Medical Laboratory Technology, Faculty of Applied
Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
6
Department of Biology, College of Science, Princess Nourah bint
Abdulrahman University, Riyadh 11474, Saudi Arabia
7
Department of Zoology, College of Science, King Saud University,
P.O. Box 2455, Riyadh 11451, Saudi Arabia
8
Pharmacology Department, Faculty of Veterinary Medicine, Suez
Canal University, Ismailia 41522, Egypt
https://doi.org/10.1007/s12035-020-02116-9
/ Published online: 15 September 2020
Molecular Neurobiology (2021) 58:281–303
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... The standard of care for AD treatment includes cholinesterase inhibitors, NMDA receptor antagonists, and monoclonal antibodies (mAbs). However, these treatments are ineffective in improving cognition, unable to change disease progression [4], limited in number of therapeutic targets [5], [6], cause severe side effects (brain swelling, microhemorrhages with mAb) [7], lack of understanding of the aging effects on AD [8], and unable to cross the blood-brain-barrier (BBB) effectively [9] to reach all affected brain areas in AD [10]. mAbs are available to lower (Aβ), but their severe side effects make the risk-benefit profile of mAbs unclear [11], [12]. ...
Article
Full-text available
Objective: Late Onset Alzheimer’s Disease (LOAD) is the most common cause of dementia, characterized by the deposition of plaques primarily of neurotoxic amyloid-β (Aβ) peptide and tau protein. Our objective is to develop a noninvasive therapy to decrease the toxic Aβ levels, using repeated electromagnetic field stimulation (REMFS) in the brain of Alzheimer’s disease patients. We previously examined the effects of REMFS on Aβ levels in primary human brain (PHB) cultures at different frequencies, powers, and specific absorption rates (SAR).PHB cultures at day in vitro (DIV7) treated with 64 MHz with a SAR of 0.6 W/Kg, one hour daily for 14 days (DIV 21) had significantly reduced (p = 0.001) levels of secreted Aβ-42 and Aβ-40 peptide without evidence of toxicity. The EMF frequency and power, and SAR levels used in our work is utilized in MRI’s, thus suggesting REMFS can be further developed in clinical settings to lower (Aβ)levels and improve the memory in AD patients. These findings and numerous studies in rodent AD models prompted us to design a portable RF device, appropriate for human use, that will deliver a homogeneous RF power deposition with a SAR value of 0.4-0.9 W/kg to all human brain memory areas, lower (Aβ) levels, and potentially improve memory in human AD patients. Methods and procedures: The research took place at the Indiana University School of Medicine (IUSM) and Purdue University Indianapolis. The first phase was done in PHB cultures at the IUSM. Through this phase, we found that a 64 MHz frequency and an RF power deposition with a SAR of 0.4-0.6 W/kg reduced the (Aβ) levels potentially impacting Alzheimer’s disease. The second phase of the project was conducted at Purdue University, we used ANSYS HFSS (High Frequency Simulation System) to design the devices that produced an appropriate penetration depth, polarization, and power deposition with a SAR of 0.4-0.9 W/kg to all memory brain areas of several numerical models. In Phase II-B will validate the device in a physical phantom. Phase III will require the FDA approval and application in clinical trials. Results: The research parameters were translated into a designed product that fits comfortably in human head and fed from an external RF source that generates an RF power deposition with a SAR of 0.4-0.9 W/kg to a realistic numerical brain. The engineering design is flexible by varying the leg capacitors of the Meander Line Antenna (MLA) devices. Thermal outcomes of the results guarantee less than 0.5 C temperature increase within one-hour time of exposure, which can be used in clinical trials for AD patients. Design parameters include dimension of the coil, the MLA structure, conducting material, and capacitance values with the produced EM fields. The flexible design was achieved by varying the additive capacitance between conductors, and via a hybrid approach integrating a birdcage with sixteen MLA. A coil antenna size within 16 cm radius and 13 cm length was achieved. A capacitance between 6.9 nF and 9.2 nF were observed when copper materials with 16 conductors were used to achieve the research parameters in focus. Conclusion: The medical project proposed here has three phases: The initial phase of determining the research parameters for reducing Aβ levels in human brain cultures and animal studies was completed at the IUSM. The translational engineering design of the REMS device and the numerical head and Antenna devices was successfully completed and presented in this paper by Purdue University and IUSM. Future phases will require manufacture and experimental validation of the REMS device with FDA approval for human application. Clinical impact: Our biological studies in human brain cultures showed that an RF power with a SAR of 0.4-0.9 W/kg at 64 MHz, lowered Aβ levels, which potentially will prevent the death of the brain neurons and improve memory in AD. The fact that we found a safe RF power deposition with a SAR value associated with the proposed biological effects in human neurons and that 64 MHz provides a penetration depth of 13.5 cm that reaches all memory areas in a human brain makes the design and manufacture of this device of high clinical impact in the study of these exposures on the treatment of Alzheimer’s and other protein associate diseases. Also, 64 MHz and RF power deposition with similar SAR levels are administer routinely in routine MRI for more than 4 decades makes it a safe framework for these novel therapeutic strategy. Clinical and Translational Impact Statement: The basic science work presented previously is both mechanistic and translational, and would advance the field of neuroscience as well as AD. This prompted us to joint efforts between the Indiana University School of Medicine and the electrical and computer engineering at Purdue University to design and develop a suitable EMF device for human treatments. Recently, our engineering team designed a birdcage antenna that generate a homogeneous RF power deposition with the same SAR values of our biological experiments in a realistic numerical human brain. Here, the engineering research has been extended to investigate the design of a portable flexible birdcage antenna that will enable adjustments to fit physical patient’s characteristics, such as geometry, head size, and tissue dimensions. This new device is expected to improve SAR uniformity and may reduce the likelihood of untreated regions in the brains of patients during treatments. Also, here we determined that the maximum temperature rise of these exposures was less than 0.5◦C, which is a safe level per regulatory agencies. This study considers a portable device system that will achieve the research parameters and patient satisfaction regarding reliability and comfort.
... In the field of advanced drug discovery, it is natural to think about multi-target compounds [37,38]. The multi-target approach was proposed almost thirty years ago when a group of researchers proved that it would be possible to design a compound endowed with a multi-target mechanism of action [39][40][41]. Over the years, this strategy has been applied to several diseases, such as inflammation, cancer and, in particular, Alzheimer's diseases (AD) [42][43][44][45]. ...
Article
Full-text available
In modern medicinal chemistry, drug discovery is a long, difficult, highly expensive and highly risky process for the identification of new drug compounds [...]
... Only five medications have received FDA approval to treat AD thus far. Four of these five medications-donepezil, galantamine, rivastigmine, and tacrine-are AChE inhibitors (AChEIs), and memantine is the only NMDAR antagonist (as shown in Figure 3) that has been approved [27][28][29][30][31][32]. Memantine, which is an NMDA receptor antagonist, seeks to control excitotoxicity while giving only symptomatic relief [33]. ...
Article
Full-text available
Gamma-glutamate is an important excitatory neurotransmitter in the central nervous system (CNS), which plays an important role in transmitting synapses, plasticity, and other brain activities. Nevertheless, alterations in the glutamatergic signaling pathway are now accepted as a central element in Alzheimer’s disease (AD) pathophysiology. One of the most prevalent types of dementia in older adults is AD, a progressive neurodegenerative illness brought on by a persistent decline in cognitive function. Since AD has been shown to be multifactorial, a variety of pharmaceutical targets may be used to treat the condition. N-methyl-D-aspartic acid receptor (NMDAR) antagonists and acetylcholinesterase inhibitors (AChEIs) are two drug classes that the Food and Drug Administration has authorized for the treatment of AD. The AChEIs approved to treat AD are galantamine, donepezil, and rivastigmine. However, memantine is the only non-competitive NMDAR antagonist that has been authorized for the treatment of AD. This review aims to outline the involvement of glutamate (GLU) at the molecular level and the signaling pathways that are associated with AD to demonstrate the drug target therapeutic potential of glutamate and its receptor. We will also consider the opinion of the leading authorities working in this area, the drawback of the existing therapeutic strategies, and the direction for the further investigation.
... Current AD therapeutics utilizing MTDs, such as AChE inhibitors and NMDA receptor antagonists, are expected to provide a more effective approach by binding to various enzymes and receptors. New compounds based on 1,2,4-thiadiazole combined with tacrine demonstrate efficient inhibition of ChEs, particularly exerting a dominant effect on BuChE, while simultaneously blocking two binding sites on the NMDA receptor [197]. Caproctamine is a typical example of MTDs which has synergistic cholinergic action against AD. ...
Article
Full-text available
Alzheimer's disease (AD), a chronic neurodegenerative disease, is clinically characterized by loss of memory and learning ability among other neurological deficits. Amyloid plaques, hyperphosphorylated tau protein, and neurofibrillary tangles involve in AD etiology. Meanwhile, enzymes and their inhibitors have become the focus of research in AD treatment. In this review, the molecular mechanisms involved in the pathogenesis of AD were overviewed and various enzymes such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), β-secretase, γ-secretase, monoamine oxidase (MAO), and receptor of advanced glycation end products (RAGE) were highlighted as potential targets for AD treatment. Several hybrid molecules with essential substructures derived from various chemotypes have demonstrated desired pharmacological activity. It is envisioned that the development of new drugs that inhibit enzymes involved in AD is a future trend in the management of the disease.
Article
Our study examines the potential of flavonoid-rich extracts from avocado seeds (Persea americana) in combating Alzheimer’s disease and diabetes. The antidiabetic activity of the extracts was assessed via the inhibition of α-amylase and α-glucosidase enzymes. Additionally, their anti-Alzheimer properties were evaluated through their anti-cholinesterase activities against AChE, BChE, and monoamine oxidase (MAO). Molecular docking and dynamics simulations were conducted to identify potential flavonoid inhibitors for the enzymes α-amylase, α-glucosidase, AChE, BChE, and monoamine oxidase. Notably, the extracts exhibited significant inhibitory activity against AChE (IC50 = 38.105 ± 0.32 µg/mL) and BChE (IC50 = 72.542 ± 1.470 µg/mL). MAO suppression protected against Fe2 + -mediated brain damage. Additionally, the flavonoid-rich extract of P. americana showed considerable inhibitory activity against α-amylase (IC50 = 608.516 ± 26.917 μg/mL) and α-glucosidase (IC50 = 790.570 ± 6.846 μg/mL) activities. HPLC–DAD profiling revealed the presence of gallic acid, caffeic acid, ferulic acid, rutin, p-coumaric acid, and quercetin. Molecular docking studies identified rutin, ferulic acid, and quercetin as the most promising ligands for the five protein targets investigated. Molecular simulations confirmed the stability of the protein‒ligand complexes, as evidenced by favorable thermodynamic parameters. Overall, our findings revealed that P. americana seed extracts have promising anti-Alzheimer’s and antidiabetic effects.
Article
Full-text available
Alzheimer's disease (AD) is a significant neurodegenerative disorder primarily affecting individuals over the age of 65. It is characterized by impairments in memory, thinking, analytical judgment, visuospatial recognition, and mood. In recent years, the development of protein and peptide drugs targeting amyloid‐beta (Aβ) has gained momentum, with several therapies entering clinical trials and even receiving marketing approval. Novel functional protein and peptide drugs, as the first‐generation immunotherapeutic agents for neurodegenerative diseases, have pioneered cellular immunotherapy for AD. However, the currently available drugs are associated with toxicity issues, which can lead to serious complications such as cerebral hemorrhage or edema. Consequently, this study examines the potential for a new generation of Aβ‐targeting drugs to mitigate the side effects of existing treatments and offers innovative perspectives for the advancement of therapies for AD.
Article
Full-text available
Autophagy is an intracellular catabolic system by which cells degrade and recycle cytoplasmic constituents such as organelles and proteins through the lysosome-dependent path. Belgian biochemist, Christian de Duve coined the name "autophagy" in 1963. The identification of autophagy-related genes and finally discoveries of mechanisms for autophagy led to the award of the 2016 Nobel Prize in Medicine or Physiology to Japanese cell biologist, Yoshinori Ohsumi specializing in autophagy. Autophagy shows a crucial role in removing aggregated proteins and damaged organelles in order to conserve intracellular homeostasis. In order to control cellular homeostasis and disease states autophagy illustrates a vital role. For insulin resistant patients, insulin secretion and the mass of pancreatic β-cells is increased on account of alterations in the expression and activities of numerous proteins in β-cells. Simultaneously, autophagic activity seems to also be upgraded to adjust to the dynamic changes taking place in β-cells. Actually, faulty autophagy in β-cells recaps a number of features that are perceived in pancreatic islets in the course of the formation of type 2 diabetes mellitus (T2DM). In contrast, the dyregulation of autophagic function is also appears to happen in the β-cells of T2DM patients. Furthermore, autophagy deficiency is linked with the diabetes-related organ dysfunction. Therefore the intention of this study was to provide the impacts of autophagy based on current researches in the development of T2DM and to explore new therapeutic strategies for stopping T2DM pathogenesis.
Article
Full-text available
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder related to age, characterized by the cerebral deposition of fibrils, which are made from the amyloid-β (Aβ), a peptide of 40–42 amino acids. The conversion of Aβ into neurotoxic oligomeric, fibrillar, and protofibrillar assemblies is supposed to be the main pathological event in AD. After Aβ accumulation, the clinical symptoms fall out predominantly due to the deficient brain clearance of the peptide. For several years, researchers have attempted to decline the Aβ monomer, oligomer, and aggregate levels, as well as plaques, employing agents that facilitate the reduction of Aβ and antagonize Aβ aggregation, or raise Aβ clearance from brain. Unluckily, broad clinical trials with mild to moderate AD participants have shown that these approaches were unsuccessful. Several clinical trials are running involving patients whose disease is at an early stage, but the preliminary outcomes are not clinically impressive. Many studies have been conducted against oligomers of Aβ which are the utmost neurotoxic molecular species. Trials with monoclonal antibodies directed against Aβ oligomers have exhibited exciting findings. Nevertheless, Aβ oligomers maintain equivalent states in both monomeric and aggregation forms; so, previously administered drugs that precisely decrease Aβ monomer or Aβ plaques ought to have displayed valuable clinical benefits. In this article, Aβ-based therapeutic strategies are discussed and several promising new ways to fight against AD are appraised.
Article
Full-text available
Alzheimer’s disease (AD) is a multifactorial and chronic neurodegenerative disorder that interferes with memory, thinking, and behavior. The consumption of dietary fat has been considered a vital factor for AD as this disease is related to blood-brain barrier function and cholesterol signaling. The ε4 allele of apolipoprotein E (APOE4) is a primary genetic risk factor that encodes one of many proteins accountable for the transport of cholesterol and it is deemed as the leading cholesterol transport proteins in the brain. In case of AD development, the causative factor is the high level of serum/plasma cholesterol. However, this statement is arguable and, in the meantime, the levels of brain cholesterol in individuals with AD are extremely inconstant and levels of cholesterol in the brain and serum/plasma of AD individuals do not reflect cholesterol as a risk factor. In fact, APOE4 is neither fundamental nor sufficient for the advancement of AD; it just acts as a synergistic and increases the danger of AD. Another noticeable characteristic of AD is area-specific decreases in the metabolism of brain glucose. It has been found that the brain cells cannot efficiently metabolize fats; hence, they totally rely upon glucose as a vitality substrate. Thus, suppression of glucose metabolism can possess an intense effect on brain actions. Hypometabolism is frequently found in AD and has quite recently achieved impressive consideration as a plausible target for interfering in the progression of the disease. One promising approach is to keep up the normal supply of glucose to the brain with ketone bodies from the ketogenic diet signifies a potential therapeutic agent for AD. Therefore, this review represents the role of ketogenic diets to combat AD pathogenesis by considering the influence of APOE.
Article
Full-text available
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that causes chronic cognitive dysfunction. Most of the AD cases are late onset, and the apolipoprotein E (APOE) isoform is a key genetic risk factor. The APOE gene has 3 key alleles in humans including APOE2, APOE3, and APOE4. Among them, APOE4 is the most potent genetic risk factor for late-onset AD (LOAD), while APOE2 has a defensive effect. Research data suggest that APOE4 leads to the pathogenesis of AD through various processes such as accelerated beta-amyloid aggregations that raised neurofibrillary tangle formation, cerebrovascular diseases, aggravated neuroinflammation, and synaptic loss. However, the precise mode of actions regarding in what way APOE4 leads to AD pathology remains unclear. Since APOE contributes to several pathological pathways of AD, targeting APOE4 might serve as a promising strategy for the development of novel drugs to combat AD. In this review, we focus on the recent studies about APOE4-targeted therapeutic strategies that have been advanced in animal models and are being prepared for use in humans for the management of AD.
Article
Neurochemistry is the specific study of neurochemicals including neurotransmitters and other molecules such as psychopharmaceuticals and neuropeptides influence the function of neurons. This section of study determines how neurochemicals influence the network of neural operation. The brain transfers numerous chemical information via neurons to communicate. The main role of neurochemistry activities takes place in the brain, which allows it to perform numerous actions. Foundation of brain is a little bit different from man to man and several things can play a role in the levels of various neurotransmitters in the brain. It is supposed that differences in brain chemistry may accountable for a variety of behavioral disorders. A particular cell called neurons is the basis of brain. Neurotransmitters have the capability that it can trigger when ordered to do so, along with receptors for specific neurotransmitters. By sending messages with neurotransmitters to signal various cell activities, brain perform its functions. Neurotransmitter spreads chemical messages from neuron to neuron to broadcast certain work and thus it works. A neuron may accept many chemical messages, both positive and negative from the other neurons contiguous it. They are accountable to get the neuron to reply in different ways, or they may work combine to produce a certain effect. Since all of this occurs just within a split second, the neurotransmitter must be cleared away rapidly so that the same receptors can be activated again and again. Psychoactive drugs work by briefly influencing a man's neurochemistry, which thusly causes changes in a man's mind-set, cognition, perception and behavior. Neuropeptides are endogenous protein molecules that are utilized for neuronal signaling. These molecules exert more prolonged and diverse effects on behavior than neurotransmitters. Therefore the objective of this appraisal is to show study of the brain’s chemical makeup especially neurotransmitters, psychopharmaceuticals, neuropeptides and their activities to nervous tissue.
Article
Neurodegenerative diseases (NDs) are characterized by disorders with progressive deterioration of the structure and/or function of neurons. Genetic mutations can lead to many NDs. Nevertheless, neurodegeneration can also take place due to several biological processes. The pathogenesis of several NDs including Alzheimer's (AD), Parkinson's (PD), and Huntington's (HD) diseases are associated with oxidative stress (OS). In order to maintain the normal functions of neurons, lower levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important, since their increased levels can cause neuronal cell death. It has been found that OS-mediated neurodegeneration involves a number of events including mitochondrial dysfunction, Ca2+ overload, and excitotoxicity. A growing number of studies are suggesting the benefit of using polyphenols for the treatment of neurodegenerative disorders. Indeed, in order to treat most of the NDs, synthetic drugs are extensively used which are found to exert side effects in the course of the treatment. There is mounting evidence that researchers have identified several naturally-occurring chemical compounds in plants, which are used for the management of NDs. Overall, polyphenolic phytochemicals are safer in nature and have negligible side effects. In this article, we have focused on the potential efficacy of polyphenols such as epigallocatechin-3-gallate, curcumin, resveratrol, quercetin and methylated polyphenols berberine against the most common neurodegenerative disorders.
Article
Alzheimer's disease (AD) is a progressive neurodegenerative disease that causes problems with memory, thinking, and behavior. Currently, there is no drug that can reduce the pathological events of this degenerative disease but symptomatic relief is possible that can abate the disease condition. N-methyl-D-aspartate (NMDA) receptors exert a critical role for synaptic plasticity as well as transmission. Overstimulation of glutamate receptors, predominantly NMDA type, may cause excitotoxic effects on neurons and is recommended as a mechanism for neurodegeneration. Atypical activation of the NMDA receptor has been suggested for AD by synaptic dysfunction. NMDA receptor antagonists especially memantine block the NMDA receptor and can reduce the influx of calcium (Ca2+) ions into neuron, thus, toxic intracellular events are not activated. This review represents the role of NMDA receptors antagonists as potential therapeutic agents to reduce AD. Moreover, this review highlights the repositioning of memantine as a potential novel therapeutic multitargeting agent for AD.
Article
Amyloid beta (Aβ) is an intricate molecule that interacts with several biomolecules and/or produces insoluble assemblies and eventually the nonphysiological depositions of its alternate with normal neuronal conditions leading to Alzheimer’s disease (AD). Aβ is formed through the proteolytic cleavage of the amyloid precursor protein (APP). Significant efforts are being made to explore the exact role of Aβ in AD pathogenesis. It is believed that the deposition of Aβ in the brain takes place from Aβ components which are derived from the brain itself. However, recent evidence suggests that Aβ derived also from the periphery and hence the Aβ circulating in the blood is capable of penetrating the blood-brain barrier (BBB) and the role of Aβ derived from the periphery is largely unknown so far. Therefore, Aβ origin determination and the underlying mechanisms of its pathological effects are of considerable interest in exploring effective therapeutic strategies. The purpose of this review is to provide a novel insight into AD pathogenesis based on Aβ in both the brain and periphery and highlight new therapeutic avenues to combat AD pathogenesis.
Article
Neurodegenerative disorders present with progressive and irreversible degeneration of the neurons. The Alzheimer’s disease (AD) affected 50 million people worldwide in 2017, which is expected to be double every 20 years. Primarily affected by age, AD is the cause for old-age dementia, progressive memory loss, dysfunctional thoughts, confusion, cognitive impairment and personality changes. Neuroglia formerly understood as “glue” of the brain neurons consisting of macroglia (astrocytes, and oligodendrocyte), microglia and progenitors NG2-glia, constitute a large fraction of the mammalian brain. The primary functions of glial cells is to provide neurons with metabolic and structural support in the healthy brain, however they attain a “reactive” state from the “resting” state upon challenged with a pathological insult such as a neurodegenerative cascade. Failure or defects in their homeostatic functions (i.e., concentration of ions, neurotransmitters, etc.), ultimately jeopardizes neurons with excitotoxicity and oxidative stress. Moreover, the most common clinical outcome of AD is the cognitive impairments and memory loss, which are attributed mainly by the accumulation of Aβ. Failure of glial cells to removes the Aβ toxic proteins accelerates the AD progression. The rapidly emerging proteomic techniques such as mass spectrometry (MS), Cross-linking mass spectrometry, Hydrogen deuterium trade mass spectrometry, Protein foot printing, 2-DGE combined with LC–MS/MS present wide array of possibilities for the identification of differentially expressed proteins in AD.
Article
Objective: Alzheimer's disease (AD) is a devastating neurodegenerative disorder, characterized by the extracellular accumulations of amyloid beta (Aβ) as senile plaques and intracellular aggregations of tau in the form of neurofibrillary tangles (NFTs) in specific brain regions. In this review, we focus on the interaction of Aβ and tau with cytosolic proteins and several cell organelles as well as associated neurotoxicity in AD. Summary: Misfolded proteins present in cells accompanied by correctly folded, intermediately folded, as well as unfolded species. Misfolded proteins can be degraded or refolded properly with the aid of chaperone proteins, which are playing a pivotal role in protein folding, trafficking as well as intermediate stabilization in healthy cells. The continuous aggregation of misfolded proteins in the absence of their proper clearance could result in amyloid disease including AD. The neuropathological changes of AD brain include the atypical cellular accumulation of misfolded proteins as well as the loss of neurons and synapses in the cerebral cortex and certain subcortical regions. The mechanism of neurodegeneration in AD that leads to severe neuronal cell death and memory dysfunctions is not completely understood until now. Conclusion: Examining the impact, as well as the consequences of protein misfolding, could help to uncover the molecular etiologies behind the complicated AD pathogenesis.