Abstract
Alzheimer’s disease (AD) is characterized by the continuous decline of the cognitive abilities manifested due to the accumulation of large aggregates of amyloid-beta 42 (Aβ42), the formation of neurofibrillary tangles of hyper-phosphorylated forms of microtubule-associated tau protein, which may lead to many alterations at the cellular and systemic level. The current therapeutic strategies primarily focus on alleviating pathological symptoms rather than providing a possible cure. AD is one of the highly studied but least understood neurological problems and remains an unresolved condition of human brain degeneration. Over the years, multiple naturally derived small molecules, including plant products, microbial isolates, and some metabolic byproducts, have been projected as supplements reducing the risk or possible treatment of the disease. However, unfortunately, none has met the expected success. One major challenge for most medications is their ability to cross the blood–brain barrier (BBB). In past decades, nanotechnology-based interventions have offered an alternative platform to address the problem of the successful delivery of the drugs to the specific targets. Interestingly, the exciting interface of natural products and nanomedicine is delivering promising results in AD treatment. The potential applications of flavonoids, the plant-derived compounds best known for their antioxidant activities, and their amalgamation with nanomedicinal approaches may lead to highly effective therapeutic strategies for treating well-known neurodegenerative diseases. In the present review, we explore the possibilities and recent developments on an exciting combination of flavonoids and nanoparticles in AD.
Keywords: Flavonoids, Nanoparticles, Alzheimer’s disease, Drug delivery, Therapeutics
Alzheimer’s disease (AD) is the most prevalent form of dementia that develops through a progressive deterioration of cognitive functions of individuals of all ages. It steadily progresses from neuronal losses and brain atrophy to the reduction in quality of life, culminating in death. The precise origin of the disease pathology and subsequent death of postmitotic neuronal cells in the brain’s hippocampus and cerebral cortical areas is not very well understood. Several genetic mutations and abnormal processing of specific proteins underlie as the causative factor toward the disease initiation. The primarily described genes having close association with AD are amyloid precursor protein (APP), beta-secretase (BACE1), presenilins (PSEN1and PSEN2), and apolipoprotein E (APOE). In recent years, several genome-wide mapping studies have identified many new candidate genes that may have possible roles in disease pathology, for example, TREM2, UNC5C, ADAM10, CLU, BIN1, ABCA7, PICALM, SORL1, etc.1,2 Nonetheless, two distinct lines of hypotheses pose amyloid-beta 42 (Aβ42)-laden plaques and neurofibrillary tangles formed of hyperphosphorylated microtubular protein tau as the possible causes of the disease; however, a direct correlation between their deposition is largely missing in most of the studies done so far.3,4 Several lines of thought also consider many other factors as the possible causes of disease pathogenesis and cell death, e.g., oxidative stress and neuroinflammation that occur in many other neuronal disorders.5,6
The mechanisms of cognitive impairments are primarily known, which include widespread synaptic dysfunctions and loss of neuronal signaling in several brain areas associated with memory and behavioral attributes.7,8 Currently, available therapeutic options for treating AD are merely symptomatic and unsatisfactory due to limited clinical application attributed to a very short half-life and fast degradation of drugs.9,10 Applying higher doses to obtain optimized and consistent effects in providing relief from the pathological conditions may result in adverse systemic effects, which may include nausea, vomiting, headache, confusion, dizziness, hallucinations, and depression, along with constipation, hepatotoxicity, and diarrhea.11,12 A major challenge in effective AD chemotherapy is the accessibility of drugs across the blood–brain barrier (BBB).13 Unsuitable lipophilicity, molecular weight, or charge of drug moieties restrict the effective drug delivery and passage to the CNS through the BBB that maintains the homeostasis in the brain and restricts the diffusion of different kinds of foreign components through the blood vessels to the nervous tissues.14
Over the past decade, nanotechnological approaches to improve drugs’ administration without causing adverse drug reactions have been widely explored. Over the time, many relevant strategies have emerged that can help the drug formulation cross the BBB and manipulate or interfere with the production, aggregation, and clearance of Aβ.15 The nanoparticles (NPs) can be exercised as the carriers of pharmaceutical molecules ranging from drugs, antibodies, genes, and peptides. Several attempts of targeted drug delivery with the help of engineered NPs have been reported successfully. For instance, the development of polysorbate 80 coated poly n-butylcyanoacrylate (PBCA) and poly lactic-co-glycolic acid (PLGA) nanoparticles loaded with drug molecules like rivastigmine have shown significantly increased drug delivery to the brain with decreased side effects.16,17 When applied on tacrine, similar approaches enhanced its brain uptake by four-fold compared to the free drug.18 Encapsulated chitosan nanoparticles of rivastigmine showed enhanced drug uptake and bioavailability in the brain.19 The encapsulation in NPs can protect bioactive molecules from systemic degradation, prolong their sustenance in the circulation, enhance the bioavailability, and allow an effective passage through the BBB with minimal side effects. Drug-loaded multifunctionalized NPs can be targeted to specific diseased areas of the brain. Moreover, newly developed biocompatible nanomaterials with improved optical and magnetic properties may also act as excellent alternatives for an early disease diagnosis.20
As a plethora of changes in the aging brain may lead to multiple pathologies, an ideal chemotherapeutic agent is required to be particularly effective for blocking multiple pathways leading to AD development.21 Compounds with multitudinous biological activities, e.g., flavonoids, can possibly affect various age-associated changes in the human brain, which otherwise may contribute to disease development and progression.22 A recent study examining the relationship between intake of flavonoids and risk of AD provides convincing pieces of evidence that long-term intakes of a flavonoid-rich diet are associated with reduced risks of developing AD.23 Several other studies highlight the potential of flavonoids to reduce or prevent all of the major changes associated with the aging brain underlying the development of AD.24,25 These changes include intensified oxidative stress, altered protein processing, decrease in neurotrophic factor signaling, accompanied by synaptic dysfunction, aggressive neuroinflammation, and activation of cell death pathways, which may altogether contribute to behavioral impairments and cognitive dysfunction.26
Low bioavailability and the highly unstable nature of flavonoids obstruct their use as possible therapeutics for neurodegenerative disease treatment.27,28 Therefore, developing new generation approaches for drug administration or delivery, for example, nanomedicinal formulations, can serve better for clinical success.29 The present review discusses nanotechnology-enabled approaches that are being developed to enhance the biomedical potential of flavonoids for therapeutic intervention and AD prevention. In addition to a detailed report on causative factors and summarizing currently available nanotechnology-based drug delivery systems for AD treatment, we highlight the impact of flavonoids on various AD hallmarks to delay or suppress the progressive neuronal loss. Although the therapeutic potential of flavonoids and nanomedicine for the treatment of AD has been discussed individually several times, collective literature of amalgamation of the interface of flavonoids and nanotechnology is not available. Foreseeing the progress in nanotechnological research, we can assume that flavonoid-based nanoformulations may lead to highly effective therapeutic strategies for treating AD in the near future.
Essentials of Alzheimer’s Disease: Genetics, Etiology, Pathways, Symptoms, and Pathophysiology
Alzheimer’s disease (AD) is clinically characterized by a progressive decline in cognition and memory formation due to an irreversible loss of terminally differentiated brain cells, mainly in the cortex and hippocampus regions of the brain.30 It is an acquired disorder of behavioral impairment. Early symptoms of AD include subtle and poorly recognized failure of memory, like failing to recall current conversations, names, or events, which slowly becomes severe due to symptoms like confusion, poor judgment capabilities, noticeable language disturbance, agitation/withdrawal, hallucinations followed by difficulty in speaking, swallowing, and walking.31 Occasionally, relapsing seizures, heightened muscle tone, myoclonus, incontinence, and mutism may also occur. Death usually results from general inanition, malnutrition, and pneumonia.32 According to “World Alzheimer Report 2019”, 50 million people have been diagnosed with dementia worldwide, which could grow further up to 150 million by 2050.33 Alzheimer’s disease (AD) accounts for approximately 70% of all the dementia cases estimated worldwide.34 Given the current projections, AD will have enormous social and economic impacts in the upcoming years. Moreover, unlike other health problems, the number of deaths caused by AD has increased by 89% between 2000 and 2014.33 Therefore, in light of growing evidence of global prevalence, associated mortality, and economic cost of the disease, increased awareness, improved early diagnosis, and providing a promising cure are a top priority.35,36
Two major molecular changes characterize the onset of AD: the aggregation of β-amyloid (Aβ) peptide and the formation of neurofibrillary tangles due to tau protein hyperphosphorylation. Simultaneously, other changes such as augmented oxidative stress, mitochondrial apoptosis, inflammation, and synaptic dysfunction may occur, which gradually culminates in the death of neurons.37,38 Despite decades of research, underlying causes and mechanisms responsible for these molecular changes and AD progression in the brain tissues remain ambiguous.39,40 Several hypotheses have been proposed to explain the genetics, onset, etiology, pathogenesis, and cure of this neurodegenerative disorder. According to highly worked out amyloidopathy, the aggregation of Aβ or more specifically Aβ42 takes place by the sequential cleavage of the amyloid precursor protein (APP) by β-secretase (BACE1) and γ-secretase yielding insoluble Aβ fibrils.41 These fibrils gradually get polymerized into extracellular spaces in the form of senile plaques, which are responsible for the synaptic damage and memory deficit in AD.42
According to the second line of hypothesis, called tauopathy, the intracellular deposition of neurofibrillary tangles (NFT) is due to the rapid phosphorylation of microtubule-associated tau (τ) protein.43 These proteins, Aβ42, total-, and phospho-tau, are primary biomarkers detected in the patients’ cerebrospinal fluid (CSF) and are used in the characterization of the disease.44 In addition, several proteins could be used as possible indicators of many other pathological changes associated with AD, indicating the disease progression.45 For example, neurofilament light (Nfl) protein shows neuroaxonal degeneration, whereas neurogranin (Ng) indicates possible synaptic degeneration.46,47 The other CSF biomarkers used to predict the disease pathology are linked with the detrimental changes associated with neuroinflammation. The primary proteins representing this category are YKL-40 and GFAP for astrocytic, and TREM2 and TSPO for microglial activation.48,49 On the basis of recent meta-analyses and investigations, a number of risk factors have been outlined. They are hypertension, homocysteine levels, heightened inflammation, diabetes mellitus, and obesity.50−54 However, age remains the most crucial risk factor for AD and other disorders of the same class. Even though the disease is more prevalent among the older section of the society (late-onset), the younger population may also develop early onset symptoms of AD pathology that may occur due to various genetic mutations in the primary genes involved in APP processing, e.g., APP, BACE1, PSEN1, and PSEN2.32,55,56 Alongside, mutations in other genes, such as TREM2, clusterin, BIN1, ADAM10, and apolipoprotein E type 4 (APOEε4), are also considered the possible risk factors for AD.57,58
APP encodes transmembrane amyloid precursor-like proteins, which generates amyloidogenic fragments under disease conditions, whereas PSEN1 and PSEN2 are responsible for coding γ-secretase enzyme complex.59 Overexpression of the APP gene (on chromosome 21) in persons with trisomy 21/Down syndrome has also been found to develop the AD hallmarks after 40 years of age. The presumed reason could be the overproduction of β-amyloid in the brains of individuals with an additional copy of the gene.60 On the contrary, late-onset AD is polygenic or guided by multiple factors. Another common hypothesis that runs parallel to the theories mentioned above is the dysfunction of the cholinergic system, causing a reduction in cholinergic markers, memory deficits, and AD progression.61,62 Genetic and molecular alterations explaining this hypothesis include the presence of APOEε4 and decreased level of phospholipase A2 (PA2) protein, an enzyme responsible for conversion of phosphatidylcholine to choline in the frontal and parietal cortices of AD patients.63,64 The critical roles of exosomal transport in the progression of many neuronal disease pathologies are now widely established. The exosomes and extracellular vesicles (EVs) may play a wide variety of roles, including the spreading of molecules inducing oxidative/proteotoxic stresses and neuroinflammation.65 However, their roles in spreading amyloids, as they do in prion diseases, are reported in several studies, but it is yet to be established.66,67 In addition, the involvement of EVs in the clearance of disease-associated plaques is also an area of research. Several studies postulate that these membranous structures may play essential roles in the clearance of proteotoxic burden and transport neuroprotective substances. Therefore, the possibilities of their application in AD therapeutics are also under consideration.65,68
Inflammatory mediators may lead to the activation of microglia. The activated glial cells contribute to AD pathology by increasing the expression of inducible nitric oxide synthase (iNOS), which produces large amounts of nitric oxide that mediates glial-induced neuronal death through inhibition of mitochondrial respiration.69 Over the past few years, a novel hypothesis related to inflammation has gained public attention, which provides a connecting link between Aβ aggregation and the NFT formation.70 According to this hypothesis, increased inflammation in the brain may be responsible for an increased risk of AD. Surprisingly, consistent inflammatory response facilitates and enhances Aβ and NFT pathologies simultaneously.71 Although inflammation protects the brain from infection, injury, and foreign threats, a slight imbalance of anti-inflammatory and pro-inflammatory signaling in the brain may give rise to chronic neuroinflammation characterized by activated microglial cells and cytokines release leading to incessant immune response.72−75 Aging is typically accompanied by a shift of innate immunity toward a pro-inflammatory status, which negatively influences brain pathology.76 Previous reports suggest that upregulation of pro-inflammatory cytokine expression (such as tumor necrosis factor-alpha or interleukin-1) by Aβ and activation of astrocytes and oligodendrocytes (the most common types of cells in the brain) can altogether play leading roles in causing neuronal cytotoxicity and generation of reactive oxygen species (ROS).77
Neurons are extremely susceptible to oxidative damages due to their high metabolic activity and loss of antioxidant capacity; therefore, excessive free radical generation and mitochondrial dysfunction may collectively lead to neuronal dysfunction and degeneration.78 It is well-known that brain-derived neurotrophic factor (BDNF) is essential for cognition and memory formation. It affects synaptic plasticity, and a reduced supply of BDNF to the basal forebrain cholinergic neurons may lead to the emergence of neurodegenerative diseases.79 Substantial studies on deficient neurotrophic hypothesis correlate incidences of AD with BDNF mRNA levels, axonal transport deficits, uneven distribution, and dysregulation of neurotrophic factors.80 Furthermore, defects in the formation and trafficking of neurotrophin signaling endosomes due to increased APP levels also result in neuronal dysfunction and AD pathogenesis.81
Curing Alzheimer’s: A Glimpse of Available Therapeutics
Currently, researchers are mainly focused on drugs that can minimize the synthesis and aggregation of Aβ and/or decrease tau phosphorylation. Molecules targeting cholinesterase and preventing the formation of amyloidic aggregates in the brain tissues could be the other set of possible drug candidates.82,83 Furthermore, statins, cholesterol-lowering drugs, anti-inflammatory substances, antioxidants molecules, and antibody-based treatment plans have also been investigated thoroughly in the past few decades to lower the symptoms and improve the quality of life of the patients.84−88 Further elucidation of AD etiology to discover novel therapeutic molecules capable of blocking AD progression efficiently and developing strategies to deliver to the regions against BBB may be helpful. Numerous clinical data suggested that impairment of cholinergic–neurotransmitter systems, possibly due to the suppression of acetylcholine (crucial for neural synapse) by acetylcholinesterase (AChE) activity and activation of the glutamatergic system, also plays a very aggressive role in AD pathology.89,90 In recent years, acetylcholinesterase inhibitors (AChEI) and N-methyl-d-aspartate (NMDA) receptor antagonists are recognized as a potential therapeutic intervention to counter the disease-linked perturbances.92
The FDA-approved drugs for AD treatment include donepezil, rivastigmine, galantamine (reversible AChEIs), and memantine (NMDA inhibitor).91,92 Interestingly, the mechanism of action of these drugs are dissimilar; for example, the most prescribed and highly selective drug donepezil is a noncompetitive inhibitor of cholinesterases, whereas rivastigmine inhibits the action of both enzymes, acetyl- and butyrylcholinesterases in a pseudoirreversible manner to delay the neurotransmission in the synapses.93,94 Galantamine interferes with the enzymatic activity of acetylcholinesterase along with the allosteric potentiation of the nicotinic acetylcholine receptors to produce greater amounts of acetylcholine in the brain.95,96 On the other hand, NMDA receptor antagonists like memantine modulate the flow of glutamatergic neuronal transmission and block the adverse effects of hyperactive glutamatergic activity like impaired synaptic plasticity and damage to the neurons. All these can efficiently manage the glutamate storm in the diseased brains.97 To relieve patients from cognitive losses, several combination therapies have also been considered clinically. Unfortunately, except the combination of cholinesterase inhibitors and memantine, the majority of combinations, though successful in preclinical studies, failed to exhibit anticipated efficacy in clinical trials.98,99
Even though ChEIs are regarded as safe drug candidates, their therapeutic effects are severely compromised due to an array of severe side effects, as discussed previously (Anand and Singh, 2013). Besides, the pharmacokinetic profile of conventional drugs in the biological system has been found to be low with substantial side-effects. This could be attributed to low bioavailability, chemical nature, or volatility of the evaluated drug candidates in the biological system.100,101 High doses of drugs (e.g., rivastigmine) to obtain an optimum level in the brain may cause adverse effects.93 A short half-life of drug molecules like tacrine, galantamine, and rivastigmine is another challenging aspect that requires further consideration.102 The oral administration of rivastigmine may lead to adverse gastrointestinal reactions, which is believed to be caused partially due to sudden increase in acetylcholine in the central nervous system. To regulate plasma concentrations of acetylcholine after rivastigmine administration and uneven distribution of the drug in the bloodstream, a transdermal rivastigmine patch formulation was developed to deliver the drug through the skin into the bloodstream.103,104
Although multiple clinical trials have been conducted in the past few years with several drug candidates designed to directly or indirectly cut the amyloid plaque load, none of these trials have shown promising outcomes.101 The complexity of AD pathology, incomplete understanding of the underlying pathways involved in the development of AD, subsequent neurodegeneration, and the lack of efficacy of available agents are few probable reasons for the failure.98 The treatment of AD is particularly complicated due to the presence of the BBB, a physical and biochemical barrier protecting the brain from potentially hazardous substances in the blood flow by restricting the passage of foreign components to the central nervous system (CNS). Most of the conventional drugs fail to cross the BBB and get eliminated by high drug efflux P-glycoprotein. Moreover, drugs carbamazepine are extremely toxic, rendering the therapeutic strategies extremely ineffective.105,106
Nanomedicinal Approaches for Alzheimer’s Disease: Opportunities with Multiple Challenges
The emergence of nanoscience and engineering is a landmark endeavor in the field of medicinal chemistry. In recent years, the ability to engineer the substances and materials of specific characteristics has gained a level of maturity.107 The unique physicochemical properties, superior catalytic activities, and improved functionality of nanoparticles (NPs) over their bulk equivalent materials could be attributed to its tiny size with very large surface area, tendency to self-assemble, and quantum effects associated with these materials.108,109 The versatility of these materials in biomedical applications could be extended to their high stability in biological systems, specific adeptness of targeting tissue, ability to encapsulate drugs, improved sensitivity for imaging and visualization due to enhanced contrast, and better resistance to corrosion over time due to elevated surface to volume ratio.110,111
Nanoformulations hold great potential to improve a plethora of physical and biological properties of the drugs, which may include but are not limited to enhanced solubility, superior pharmacokinetics, better selectivity, higher efficacy, and least toxicity in comparison to traditional medicines.112,113 Coupling of nanoformulations, such as polymeric NPs, solid lipid NPs, liposomes, nanoemulsions, nanostructured lipid carriers (NLC), etc., helps in the safe delivery of drugs or bioactive moieties to specific sites of action with an improved pharmacokinetic profile causing minimal adverse drug reactions. These may further assist drugs in overcoming challenges, like passage through the BBB, overpowering gastric pH barriers, increased absorption and bioavailability, prolonged half-life, reaching at the site of action in biologically appropriate therapeutic doses.105,114 In fact, an equilibrium between the physicochemical and pharmacokinetic properties is required to be maintained to optimize the effectiveness and specificity of therapeutics toward molecular targets.
Widespread applications of nanomaterials in the treatment of both chronic and acute diseases could be attributed to the promises shown in the growing number of studies based on these NPs in diverse kinds of disease models.20,115 The enormous potential of nanomedicine in the treatment of CNS diseases has been highlighted in the past few years owing to its lipophilicity, lower molecular weight (up to 400 Da), smaller size (in the range of nanometers), appropriate charge with a higher surface-to-volume ratio, which may allow easy access to the brain through the process of simple diffusion.116,117 Many studies have been done, where nanoformulations have been prepared through polymerization, ionic gelation, coacervation, emulsion solvent evaporation, etc., to target AD pathophysiology in animal models.15,118−121 There are specific benefits associated with varying preparation methods, e.g., coating NPs with peptides, surfactants, or antibodies can enormously improve the absorption and drug delivery to the brain.122 Previously, the concentration of tacrine in the brain and other tissues was improved by nanocoating polysorbate 80, a nonionic surfactant, on the tacrine-conjugated poly(n-butylcyanoacrylate) nanoparticles.18 In another study, it was found that the chitosan NPs incorporated into rivastigmine may enhance its bioavailability and drug uptake, thus improving the efficacy.123
Surface modification of the nanoparticles tends to facilitate its permeability across BBB without any phagocytic opsonization and provide longer circulation time without unnecessary interactions during their prolonged stay in the biological system.124 Nanostructured lipid carriers (NLCs) can significantly improve the bioavailability and facilitate the administration of drugs via the intranasal route.125 Such particles can greatly enhance the biodistribution of the drugs with poor water solubility and improve the drug efficacy due to the modified administration strategies, including oral, nasal, etc.126 Interestingly, the intranasal route of administration could deliver an increased amount of drugs in the brain as compared to intravenous by accessing the olfactory route and evading the BBB.127 These drug-laden NPs have enhanced absorption and delivery to the brain with the least side effects as lower doses could also be effectively administered.128 Furthermore, rivastigmine liposomes carrying cell-penetrating peptide (CPP) have shown improved distribution and retention of the drug in the brain, with enhanced pharmacodynamic properties, including lower side effects.129 Delivering drugs like donepezil and tacrine using liposomes leads to quick absorption and considerable bioavailability of the drug.130,131
Ligand-functionalized nanoliposomes can also enhance the therapeutic effects of galantamine and rivastigmine by lowering the amyloid-beta formation and ameliorating other AD-associated pathological symptoms.132,133 Drug encapsulation by such nanocarriers could extend their systemic benefits by improving the pharmacological properties of drugs and promoting their retention, bioavailability, and increasing absorption, thus imparting the anticipated therapeutic effects.134 In addition, these drug delivery systems have very targeted evasion inside the biological system with lesser chances of random interaction and activation of molecules at unspecific targets, which helps in avoiding the accumulation of possible adverse effects, including abdominal complications, vomiting, nausea, etc., which are common in many treatment plans currently in use.112,135
Although there is a lack of comprehensive understanding of AD pathogenesis, it is clear that multiple cellular pathways are affected. Targeting a single pathway may relieve the symptoms mildly but will never lead us to a reliable cure. Also, in recent years targeted antiamyloid agents and therapies, including BACE inhibitor drug molecules and anti-Aβ42 antibodies, have failed to meet the expectations in the clinical trials.136−139 Therefore, in the past few decades, researchers are looking for the beneficial aspects of naturally occurring drug candidates, which may have antioxidative and anti-inflammatory characteristics along with the definite anti-amyloidogenic properties that can prevent aggregation of Aβ42 peptides.140 Concurrently, it should have high bioavailability, water-solubility, BBB access, with a considerable half-life, absorption, and appropriate distribution in the neuronal cells. Also, lesser toxicity is another primary attribution that the natural molecules provide. Therefore, a thoughtful reconsideration of the amalgamation of natural compounds containing multiple neuroprotective properties as one resort and nanobased approaches to facilitate their delivery to specific brain regions could be a possible futuristic approach in the AD drug development pipeline.
Advantageous Therapeutic Benefits of Flavonoids Against Alzheimer’s disease
Flavonoids are polyphenolic compounds naturally present as colored pigments in higher plants.141 They are synthesized from the amino acid phenylalanine by the phenylpropanoid pathway and are ubiquitously found in fruits, vegetables, nuts, seeds, stems, flowers, and tea.142 The canonical structure of flavonoids, with more than 10 000 discovered so far, consists of three aromatic rings lying adjacent to each other.143,144 Two rings named A and B collectively form the 15 carbon flavan nucleus that is further linked with a three-carbon heterocyclic ring C.145,146 On the basis of the structure, flavonoids have been classified into six broad categories. They are flavanols, flavonols, flavones, flavanones, isoflavones, and anthocyanidins.147 Flavonoids have been primarily explored for their cytoprotective, antiviral, antiallergic, anti-inflammatory, antidiabetic, antiobesity, antitumor, and neuroprotective properties, and thus are considered as molecules of significant medicinal importance as they exhibit tremendous opportunities to explore and exploit their pharmacological properties.142,148−150
As discussed before, AD pathology is a clinical manifestation of multiple pathways altered due to loss or gain-of-functions of one or more genes, causing a rise in oxidative stress, weaker neurotrophic support, altered energy metabolism, deposition of neurofibrillary tangles, and plaque aggregation followed by inflammation and cellular damage in the specific brain regions.151,152 The plethora of AD brain changes put a challenge before researchers and clinicians to address them simultaneously; however, our limitations in understanding the primary cause of disease onset further toughens the chances of a finding possible cure soon. However, therapies directed to delay the symptoms and provide symptomatic relief are the current paradigm of AD treatment. Countering multiple age-associated changes in the brain require a combination of drugs simultaneously directed against wide range targets. Flavonoids provide a safer alternative group of compounds, which have been explored widely. Further research is continuously encouraging the idea that some of the dietary flavonoids may have a wide range of therapeutic benefits.
Figure 1.
Flavonoids: A Brief Introduction. (A) Backbone structure of flavonoids consisting of an aromatic ring A fused with an oxygenated heterocyclic ring C further connected to another aromatic ring B. (B) Pharmacological properties and roles of flavonoids in prevention of multiple pathological conditions. (C) Classification and example of flavonoids with their chemical structures. (D) Major plant parts from which flavonoids are extracted and some well-known examples of flavonoids purified from these sources.
Administration of rats with a citrus flavanoglycone hesperidin also resulted in cognitive protection against l-methionine induced oxidative stress and neurotoxicity.153 In a mammalian animal model, treatment with (−)-epigallocatechin-3-gallate (EGCG) has shown protective effects against ischemia-induced toxicity in hippocampal neurons.154 It has also shown beneficial effects in Parkinson’s disease (PD) and amyotrophic lateral sclerosis (ALS) mice models.155,156 Quercetin interferes with the enzyme proline hydroxylase (PHD), which facilitates the ubiquitination of hypoxia-inducible factor-1 alpha (HIF-1α) by the protein of von Hippel–Lindau, an E3 ubiquitin ligase, and thus stabilizes it under normal oxygen condition.157 Similarly, another transcription factor associated with redox homeostasis, nuclear factor erythroid 2-related factor 2 (Nrf2-), is reported to be activated by multiple flavonoids, e.g., pinocembrin, eriodictyol, and naringenin, under oxidative stress like conditions.158−160 Not only the suppression of the oxidative damage but getting better of the microglia- and astrocyte-mediated neuroinflammation is another possible mechanism that flavonoids have primarily been tested for their potential as anti-inflammatory neuroprotective agents.
Previous studies suggest that the downstream effects of activation of microglia-mediated inflammatory changes in the central nervous system (CNS) can initiate a detrimental chain of events leading to the death of neurons. Therefore, the positive effects of many classes of small natural molecules over neuroinflammation have been investigated in the past.161 The major flavonoids with neuroinflammation modulatory potential are baicalein, catechin, EGCG, fisetin, genistein, quercetin, and wogonin; however, they may have different mechanisms of action.162−166 Interestingly, flavonoids can regulate inflammatory events by decreasing the level of prostanoids and leukotriene with the help of eicosanoid generating enzymes like phospholipase A2, cyclooxygenases, and lipoxygenases.167 Other possible mechanisms could be the regulation of the cellular metabolic alterations by regulating multiple kinase pathways altered under various disease-associated stress conditions.168 Some flavonoids, like baicalein, epicatechin, kaempferol, and quercetin, also suppress c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinases (ERK)-dependent apoptotic pathways to protect neurons against multiple kinds of stresses and injuries.169−171
Growing evidence reinforces the notion that flavonoids play active roles in slowing down cognitive decline and dementia.172−174 Their inherent potential to subdue the progression of neurological disorders has gained special attention in recent years. The elemental role of flavonoids in stimulating cognitive functions, learning, and memory is apparently associated with their ability to induce neuronal regeneration, encourage neurogenesis, antioxidant capabilities, and protect vulnerable neurons through interaction with the molecular counterparts of the brain and intracellular neuronal pathways.175,176 Multiple lines of investigations proposed different mechanisms to explain the efficacy of flavonoids in learning and memory, clearly highlighting their potential as a possible remedy to cut down the disease severity in the patients.177 Fisetin, a natural polyphenol, can restore the long-term potentiation (LTP) by inducing ERK signaling and inducing phosphorylation of cAMP response element-binding protein (CREB) in rat hippocampi.178 Another potent memory enhancer flavonoid is luteolin, which improves the synaptic transmission and, via activation of CREB, stabilizes memory.179
A wide range of flavonoids has been reported to avert cognitive impairments and inhibit AD development through anti-amyloidogenic properties.180,181 Interestingly, multiple flavonoids have shown direct or indirect anti-amyloidogenic potential in various in vitro model systems. The most studied and widely explored anti-amyloidic flavonoids are didymin, myricetin, morin, prunin, poncirin, quercetin, and rosmarinic acid.182−184 More interestingly, in a cell system based study, EGCG has shown enhancing the release of soluble nonamyloidogenic form of APP in protein kinase C (PKC) dependent mechanism.185 Multiple lines of studies have proposed flavonoid-mediated reduction in secreted Aβ levels and inhibition of BACE-1 activity in primary cortical neurons or involvement in tau phosphorylation through suppression of the activities of several kinases including GSK-3β (tau phosphorylation) and interaction with various neuronal signaling pathways through various protein kinase- and lipid kinase-signaling cascades.186−189 Luteolin and diosmin administered mice models of AD pathology showed inhibition of GSK-3 and altered processing of presenilin-1, resulting in reduced amyloid formation and thus improve the disease condition.190
Figure 2.
An overview of Alzheimer’s disease pathophysiology: Visible morphological atrophy in the brain resulting from a cascade of events triggered by various genetic and environmental factors leading to Alzheimer’s disease. Major AD hallmarks include amyloid plaques, neurofibrillary tangles, neuroinflammatory changes, and microglial activation. Several therapeutic strategies have been proposed in the past to prevent disease progression and relieve the symptoms.
Inhibition of acetylcholine is one of the preliminary changes in AD pathology; therefore, inhibition of AChE has been one primary drug development strategy for a long time. Several independent research groups have identified a large number of flavonoids with AChE inhibition potential; tiliroside, quercetin, and rutin are prominent natural molecules of this class.191,192 Apart from these, genistein and silibinin are other flavonoids with strong in vitro inhibitory potential against both AChE and BChE.193 Multiple in silico studies have identified similar binding and inhibition of both the cholinesterases by galangin, kaempferol, quercetin, myricetin, and morin.194,195 Several flavonoids have been found upregulating the secretion of different types of neurotrophic factors in the brain cells; for example, 7,8-dihydroxyflavone (7,8-DHF) is a potent agonist of tropomyosin-receptor-kinase B (TrkB), a receptor for the most abundant neurotrophin, brain-derived neurotrophic factor (BDNF). Therefore, it may compensate for the loss of the BDNF in AD pathology by improving the branching and synapse formation by alleviating Aβ deposition and thus rescues 5xFAD mice from memory loss.196,197 We have summarized available research outcomes in the form of Table 1 that reflects the therapeutic potential of various flavonoids.
Table 1. Flavonoids and Their Therapeutic Applications in CNS.
flavonoid | model | biological target | therapeutic effect | ref |
---|---|---|---|---|
EGCG | TgCRND8 mice | soluble Aβ | improved learning and memory | (264) |
SAMP8 mice | Aβ1–42, BACE-1, tau, | rescued spatial learning | (265) | |
APPsw mice | Aβ1–42, tau | cognitive benefits | (266) | |
CUMS rats | Aβ1–42, mTOR, p70S6K | enhanced spatial learning and memory | (267) | |
anthocyanins | APdE9 mice | APP processing | alleviated behavioral and memory deficits | (268) |
curcumin | THP-1 cells | Egr1, TNFα, IL1β, MAPK | ameliorate inflammation | (269) |
PC12 cells | Aβ42 | protection from Aβ insult | (270) | |
nobiletin | APP-SL7–5 mice | Aβ42, ERK | improves memory impairment | (271) |
3xTg-AD | Aβ1–40, ROS | reversal of impaired learning and memory | (272) | |
SAMP8 mice | p-tau, SOD1, GPx | ameliorates learning and memory deficits | (273) | |
AD rats | Aβ42, PKA, CREB | rescues memory deterioration | (274) | |
resveratrol | AD patients | Aβ40, MMP9, IL-4, FGF-2 | inhibits inflammation and cognitive decline | (275) |
baicalein | Tg2576 mice | APP | improved cognitive performance | (276) |
myricetin | AD rats | AMPK, SIRT1 | suppresses apoptosis, improves learning | (277) |
quercetin | 3xTg-AD | Aβ42, tau | improves learning performance | (278) |
3xTg-AD | Aβ42, BACE1 | strengthens spatial memory | (279) | |
kaempferol | AD rats | SOD, GSH, TNFα, MDA | improves spatial learning and memory | (280) |
silibinin | Aβ treated rats | BDNF/TrkB, autophagy | ameliorates anxiety/depression behaviors | (281) |
apigenin | APP/PS1 mice | APP, BACE1, SOD1, GPx | relieves learning and memory impairment | (282) |
rutin | rat model | ERK1, CREB, and BDNF | rescues amyloid induced neurotoxicity | (283) |
wogonin | 3xTg-AD mice | APP, tau, cleaved PARP | enhances neurite outgrowth and memory | (284) |
SH-SY5Y cells | APP, tau, mTOR | may improve cognitive function | (285) | |
fisetin | 2x FAD mice | Cdk5, ERK | improves learning and memory deficits | (286) |
SAMP8 mice | Arc, GFAP, p35, SAPK/JNK | delays aging and cognitive decline | (287) | |
catechin | PC12 cells | Aβ | reduces apoptosis and neurotoxicity | (288) |
7,8-DHF | 5xFAD | BACE1, Aβ, TrkB | attenuates memory deficit | (289) |
5xFAD | TrkB | restores synapses and memory | (290) | |
morin | 3xTg-AD mice | GSK3β, tau, Aβ | increased neuroprotection | (291) |
luteolin | Tg2576 mice | GSK3β, PS1, Aβ | reduces Aβ toxicity | (292) |
diosmin | Tg2576 mice | Aβ42 | reduces Aβ toxicity | (292) |
3xTg-AD mice | Aβ42, tau | suppresses inflammation and memory loss | (293) | |
diosmetin | 3xTg-AD mice | GSK3β, γ-secretase | reduces inflammation and cognitive loss | (293) |
hesperetin | Neuro-2A cells | IR, Akt, GLUT3, GLUT4 | regulates autophagy and metabolism | (294) |
hesperidin | Neuro-2A cells | IR, Akt, GLUT3, GLUT4 | regulates autophagy and metabolism | (294) |
APP/PS1 mice | iNOS, TNFα, IL1β | restores behavioral/social interaction | (295) | |
puerarin | SAD mice | GPx, SOD, MDA | regulates oxidative stress/memory decline | (296) |
SAD mice | caspase-3, p38, JNK, Bax | reduces oxidative damage and apoptosis | (297) | |
APP/PS1 mice | Nrf2, HO1, Akt, GSK-3β | alleviates cognitive impairment | (298) | |
naringenin | rat model | GSK-3, PPARγ, IR, Tau, Aβ42 | facilitates spatial learning and memory | (299) |
naringin | AD rats | Ach, IL1β, mitochondria | mitigates cognitive decline | (300) |
genistein | SAD rats | APP, Aβ42, tau | restores autophagy, behavioral normalcy | (301) |
Advances in Nanodrug Delivery Systems and Their Benefits in the Context of Flavonoids
Flavonoids have the capability to target multiple pathways associated with several complex neuropathological conditions. Despite multifaceted benefits and very few known side-effects at the prescribed doses, they are not yet considered a suitable class for drug development, owing to their inherent instability, inefficient delivery models, and low retention in target tissues. These limitations of flavonoids have been addressed thoroughly in the past few years to attain clinical success. Nanobased formulations of many standard drugs have been successfully applied to counter the challenges and abolish the major pharmacokinetic barriers. The advanced nanotechnological approaches have shown a vast potential to narrow down the gaps between the physiological effects of the drug molecules and their possible applications in therapeutics. Many flavonoid-based nanoformulations have been prepared and examined for the promising results on cell-based systems and animal models in recent years. A detailed overview of some major studies with such nanomedicinal strategies explored widely in the context of AD is presented in Table 2.
Table 2. Advantages of Flavonoid Laden Nanoformulation against Alzheimer’s Disease Pathology.
flavonoids | experimental model | pharmacological advantages | biological activity | ref |
---|---|---|---|---|
Liposomes | ||||
baicalein | SHSY5Y, PC12 cells | slow release, high internalization | protects against oxidative stress | (205) |
resveratrol | NIH-3T3 | improved solubility | presents no cytotoxicity | (302) |
quercetin | rat model | enhanced solubility | anxiolytic and cognitive benefits | (303) |
SK-N-MC cells | enhanced access through BBB | suppression of neuronal apoptosis | (201) | |
EGCG | N2a cells, rats | increased oral bioavailability | enhances α-secretase activity | (304) |
Silica Nanoparticles | ||||
catechin | hippocampal cells | high solubility | enhances cell survival | (305) |
chrysin | hippocampal cells | improved drug entrapment | suppresses Aβ toxicity | (306) |
quercetin | hippocampal cells | retains physicochemical integrity | improves antioxidant activity | (307) |
naringin | hippocampal cells | entraps higher drug doses | reduces Aβ toxicity | (231) |
Selenium NPs | ||||
EGCG | PC-12 cells | high affinity for Aβ | disaggregates Aβ fibrils | (206) |
curcumin | 5x FAD mice | increased binding with plaques | inhibits Aβ aggregation | (308) |
Polymeric NPs | ||||
EGCG | APP/PS1 mice | increases drug stability | increased synapses | (309) |
EGCG | rats | superior bioavailability of drug | improves neurobehavioral deficits | (310) |
anthocyanin | SH-SY5Y cells | slow and sustained drug release | abrogates oxidative stress | (243) |
quercetin | SH-SY5Y cells | sustainable release | disassembles Aβ fibrils | (311) |
curcumin | SK-N-SH cells | increased solubility and stability | suppresses ROS and Nrf2 | (312) |
Lipid Core Nanocapsules | ||||
resveratrol | hippocampal cultures | restricts drug photosenstivity | reduces Aβ induced inflammation | (313) |
Protein Nanoparticles | ||||
quercetin | SAMP8 mice | greater bioavailability | improved cognition and memory | (255) |
Solid Lipid Nanoparticles | ||||
curcumin | 5× FAD mice | enhanced affinity to Aβ-plaques | may interfere with Aβ fibrillation | (212) |
resveratrol | in vitro BBB model | enhanced cellular uptake | inhibits amyloid beta aggregation | (314) |
chrysin | rats | improved bioavailability | mitigates Aβ induced stress | (315) |
quercetin | rats | higher drug entrapment | improved memory retention | (249) |
Danio rerio | improved drug release | attenuates cognitive impairments | (316) | |
Nanostructured Lipid Carriers | ||||
baicalein | rat model | higher drug accumulation | superior brain targeting | (317) |
quercetin | Caco-2 cells | increased drug residence | improved stress resistance | (318) |
Metal Nanoparticles | ||||
curcumin | Tg2576 mice | enhanced affinity to Aβ-plaques | may interfere with Aβ fibrillation | (319) |
neuro2a cells | high solubility and Aβ affinity | dissolves amyloid fibrils | (320) | |
anthocyanins | AD mice model | better release and bioavailability | inhibits neuronal apoptosis | (204) |
AD mice model | instability, bioavailability | lowers Aβ induced inflammation | (321) | |
quercetin | rat model | high plasma concentration | prevent neural cell apoptosis | (227) |
SH-SY5Y cells | high BBB permeability | enhances autophagy clearance | (322) |
We can understand this amalgamation of natural drugs and nanomedicine by a few studies performed in recent years, which have successfully addressed the clinical challenges against many such flavonoid based drug molecules. Naringenin protects neurons against the free radicals and suppresses the inflammation but has limited permeability across membranes; however, naringenin nanoemulsions delivered via a intranasal route have shown promising effects in Alzheimer’s and Parkinson’s disease models.198,199 Similarly, the neuroprotective potential of highly active quercetin could be masked by constraints, such as low solubility, fast metabolism in the system, and low accumulation in the brain.200 However, a quercetin-coated liposomal formulation was found to permeate BBB effectively and protect cellular AD models from Aβ-induced toxicity.201 Selective targeting by conjugating liposomal surface to specific receptors, e.g., lactoferrin, could be another novel strategy, which can specifically target bioactive molecules, e.g., quercetin, to the brain and enhance its accumulation through the receptor-mediated transcytosis.201 Another example of a receptor-mediated permeation approach was developed through bradykinin B2 receptor agonist RMP-7, which increases the access of hydrophilic drugs across the tight junction of the BBB.202
Anthocyanins are very unstable at gastrointestinal pH; therefore, their encapsulation in nanoemulsions has displayed increased retention with an estimated half-life of 385 days, enhancing their antioxidant potential manifold.203 Additionally, polyethylene glycol (PEG)-ylation of nanoparticles assists in preventing nonspecific interactions of flavonoids with blood proteins and promote their stability. PEGylated gold NP formulation of anthocyanin may suppress amyloid beta-induced neurotoxicity and reduce neuroinflammation through dysregulation of PI3K/Akt/GSK3β signaling and inhibition of tau hyperphosphorylation.204 Nanomodification strategies facilitate several favorable modifications to these neurologically important molecules without losing their original properties providing many advantages over traditional drug delivery schemes. A major challenge before flavonoid-based drug formulations is their sparingly soluble nature in aqueous solutions, affecting their availability in the bloodstream, followed by rapid elimination. Entrapment of baicalein, an aglycone derivative having very low aqueous solubility and stability under biological conditions, in a nanoliposomal preparation decorated with cholesterol and PEG significantly reduced its cytotoxicity and augmented its oxidative stress management, cell uptake, and neuroprotection capabilities in SH-SY5Y and PC12 cell lines.205
Increased uptake and greater accumulation of biodrugs have been achieved in recent years through several novel formulation types, e.g., solid lipid nanoparticles, nanostructured lipid carriers, and selenium nanoparticles of resveratrol, EGCG, and curcumin-like bioactive drug compounds.206−208 Theracurmin, a colloidal nanoparticle consisting of curcumin, glycerine, and a water-soluble polysaccharide, has shown encouraging outcomes in a randomized clinical study, where the solubility and oral bioavailability of curcumin was increased up to ∼16 times in comparison with unformulated curcumin when administered in healthy volunteers.209 Similarly, LongVida, a solid lipid curcumin particle (SLCP)-based formulation, improved bioavailability of curcumin in addition to protecting it from fast metabolism and elimination in osteosarcoma patients reflecting the importance of solid lipid particles in overcoming clinical constraints of traditional medicine curcumin.210 Solid lipid curcumin particles have also been designed and successfully tested against neuroblastoma cells, AD mouse and rat models, and have shown encouraging results, like improved stability and delivery across BBB.211−213
Incorporation of flavonoids to metal nanoparticles (e.g., gold) benefits maintaining the biocompatibility of NPs that ranges from a reduction in cytotoxicity caused by metal ions to preventing aggregation in an aqueous environment and providing additional stability to the nanoparticles.214 Over many years, there has been a steep increase in synthesis and development of several novel nanoformulations, which have paved the way for many drugs to be revisited and applied in neurotherapeutics. Here, we are providing a summarized overview of the specific evolution of different nanoformulations in the field of AD research.
Metallic Nanoparticles (MNPs)
Metal may induce excessive ROS production leading to an exaggeration of AD symptoms; therefore, the application of MNPs in AD pathology is debatable. Also, the accumulation of MNPs such as Ag, TiO2, ZnO, etc., in delicate organs like the brain may have profound implications leading to permanent damage.215 However, a decreased neurotoxicity has been observed in the presence of antioxidants, such as N-acetyl-l-cysteine, selenium, etc., which has neuroprotective effects.216,217 Selenium, a trace element, and its NPs have gained special attention in recent years due to its lower toxicity and antioxidant properties along with exceptional antiaggregatory properties.217,218 A formulation of selenium NPs and sialic acid-functionalized with BBB permeable peptides B6 has demonstrated exceptional results.219 Toxicity issues can also be undermined through the biological synthesis of these nanoparticles where phytochemicals and other biological sources replace harmful chemical reagents to reduce and stabilize metal ions, an approach generally referred to as green synthesis.220,221 Capping of metal nanoparticles (iron oxide, cadmium sulfide) with proteins of fungal origin also has shown disaggregation of tau without affecting the cell viability of neuroblastoma cells.222
Figure 3.
A concise overview of multifunctional nanoparticles: Schematic representation of two major approaches of NP synthesis (in the central part): Top-down and bottom-up, accomplished with the help of physical, chemical, and biological methods of synthesis (left). A representative nanoparticle is decorated with multiple ligands, antibodies, peptides, nucleic acid, etc., for targeted drug delivery (in the oval). Different types of nanoformulations commonly used for drug delivery purposes, and a few critical parameters of classifying these nanomaterials (right).
Flavonoids with immense antioxidant potential, e.g., EGCG, quercetin, anthocyanin, and curcumin, have been combined with selenium, iron oxide, and gold nanoparticles. Silver and gold NPs and various modifications of these metallic nanoparticles have been tested in multiple disease therapeutics in the past.115,223,224 Modification to these nanoscale moieties with functional groups, ligands, and macromolecules like peptides, proteins, and DNA may facilitate the greater accumulation of therapeutic substances at the target tissue.225 In a recent study, EGCG-functionalized selenium NPs were coated with Tet-1 peptides for higher cellular uptake and were able to inhibit amyloid aggregation and also to disaggregate the preformed toxic aggregates.206 Similarly, PEG stabilized gold nanorods, when dual-functionalized with Angiopep 2 and D1 peptides, had promising efficiency of reaching the CNS respectively and targeting Aβ aggregates, without compromising neuronal cell viability.226 In another study, conjugation of quercetin with super-paramagnetic iron oxide nanoparticles enhanced bioavailability and improved learning and memory in healthy rats.227
Silica-Based Nanoparticles
The therapeutic potential of silica-based nanoparticles, a promising delivery platform for targeted brain therapy, is severely compromised by its neurotoxic behavior attributed to excessive generation of oxidative stress and neuroinflammation that might increase the risk of developing AD.228,229 These adverse effects could be ameliorated by organic modification and functionalization of silica nanoparticles, consisting of stabilized monodispersed aqueous nanosuspension with surface-functionalization using amino groups.230 Silica nanoparticles synthesized as nanocarriers of flavonoids like quercetin and naringin provided beneficial results in terms of enhanced neuroprotective capacity against Aβ-induced oxidative stress. The nanoparticles were more competent in loading capacity, antioxidant activity, and release profile.231,232 A separate study involving catechin-loaded silica nanoparticles showed enhanced cell survival and protective activity in primary rat hippocampal neurons against Cu2+-induced oxidative stress.233
Liposomes
Liposomes are vesicle-like structures made up of phospholipid bilayers. They are essential in the context of AD because of their capabilities of crossing BBB and safely delivering both hydrophilic and hydrophobic molecules to their target tissues without causing much side effects.234 In some cases, liposomes can be administered intranasally, resulting in improved penetration into the brain while protecting their contents from degradation.235 Their surfaces can be modified for selective targeting using antibodies against the transferrin receptor, which increases its affinity toward the brain and amyloid-β peptides.236 They are preferred over other nanomaterials because of their biodegradability and increased cellular uptake without eliciting any immunological response.237 Liposomal formulations of curcumin have been prepared and examined both in vitro and in vivo, have shown a very high affinity for Aβ1–42 fibrils, and were significantly effective in ameliorating Aβ-induced neurotoxicity in AD mice.238,239 These formulations exhibited enhanced solubility and cellular uptake without causing toxicity. Additional PEG coating of liposomes may prolong the circulation time of the bioactive curcumin.240 In another study, liposomal formulations of quercetin showed cognitive enhancement at a much lower dose than the conventional form of the drug.241
Polymeric Nanoparticles
A number of polymeric nanoparticles in the form of either nanocapsule, where the drug molecules are enclosed inside a polymeric membrane, or nanospheres in which therapeutic agents are evenly dispersed throughout the polymer matrix, have been designed and successfully tested on AD mouse models.15 The polymer often used in such preparations PLGA can readily encapsulate hydrophobic compounds to form PLGA derived nanoparticles. These nanoparticles can be further modified to add favorable characteristics like solubility or receptor-mediated targeting of therapeutic molecules.242 For example, the efficiency of anthocyanins, curcumin, and naringin was found to be highly improved when encapsulated in PLGA or PEG alone or in combination.231,243,244 These formulations enhanced bioavailability and neuroprotective capacity of flavonoids manifolds by protecting them from degradation, oxidation, and unfavorable interactions with plasma proteins, along with their enhanced capacity of diminishing oxidative stress and destructing Aβ aggregates.
Solid Lipid Nanoparticles (SLNs)
SLNs are aqueous colloidal nanoparticles suitable for delivering both hydrophilic and lipophilic drugs to their target tissues or sites.245,246 They provide better tolerance than polymeric nanoparticles and, at the same time, appear to resolve stability and drug release issues of lipid nanoemulsions and liposomes.247 Several SLNs encapsulating chrysin and curcumin have been useful in AD animal models with increased uptake, bioavailability, permeability, and greater neuroprotection without loss in its antioxidant capabilities in rat AD models.211,248 In a different study, resveratrol has been encapsulated in an anti-transferrin receptor monoclonal antibody functionalized solid lipid nanoparticles, which has substantially enhanced uptake by human brain-like endothelial cells as compared to normal SLNs or the ones functionalized with an unspecific antibody.207 SLN-encapsulated quercetin also showed increased memory-retention in mice and attenuating effect in neurocognitive impairments along with amelioration of biochemical changes in zebrafish.249,250
Protein Nanoparticles
Protein nanoparticles could potentially transform properties of materials in terms of size and surface area, prompting them to be more reactive and available for surface modifications.251 The significance of protein-based nanostructures has recently been realized due to their wide range of applicability attributed to very high biodegradability and biocompatibility.252 A plethora of protein nanoparticles made up of proteins, like whey protein, gelatin, zein, soy protein, and milk protein, have been explored and reported by researchers.253 Flavonoids like quercetin and resveratrol encapsulated with the help of zein, a maize protein, to produce colloidal nanoparticles have been reported recently.254 Quercetin encapsulated zein nanoparticles have shown improved oral bioavailability (∼60%) with sustained plasma levels of quercetin, besides improved cognition and reduced astrogliosis in the Samp8 mice model.255 Similarly, resveratrol encapsulated zein NPs offered high and prolonged plasma levels of resveratrol with significant oral bioavailability improvement (∼50%) in mice models. Further, a pH-independent release of resveratrol from the nanoparticles and moderate augmentation in anti-inflammatory effects was observed.256 Improvements in the oral and topical bioavailability of resveratrol were also achieved with the help of encapsulated casein nanoparticles.257
Figure 4.
Therapeutic potential of flavonoid nanoformulations in Alzheimer’s disease: Combination of therapeutic properties of flavonoids with nanobased delivery systems may provide an effective therapeutic tool for the successful cure of Alzheimer’s disease, which remains one of the most challenging puzzles of the medical sciences after more than a 100 years of its discovery.
Conclusions and Future Directions
Neurodegeneration is a gradual and multifactorial process that starts with genetic or environmental factors and progresses with exacerbation of accumulated oxidative stress, proteotoxic load, compromised cellular energetics, mitochondrial dysfunction, and neuroinflammation.258,259 The growing number of evidence supports the notion that many natural molecules, especially the dietary flavonoids, could be considered therapeutic alternatives in AD due to their innumerable health benefits, including antioxidative, anti-inflammatory, and anti-amyloidogenic properties. Furthermore, flavonoids can pass through the BBB and positively support the brain from multiple kinds of stresses and damages.260 Also, their multifaceted potential of targeting different subcellular pathways and many systemic changes, like oxidative damage, glutamate storm, inflammation, and compromised proteostasis, has been explored and widely known.
Multiple studies are underway to present flavonoids as possible AD drugs at the preclinical level. Unfortunately, a few clinical studies conducted on curcumin and resveratrol in the past have failed to provide convincing results due to inefficient delivery, and some reported toxicities.261,262 In several independent studies, challenges like poor pharmacokinetic profile, low permeability across BBB, stability, and aqueous solubility for flavonoids have been observed, which may hinder or challenge these drug candidates’ potential applicability in the context of CNS disorders. However, nanobased delivery systems can greatly enhance the biomedical applications of flavonoids and other natural molecules in the context of brain diseases. The amalgamation of natural products with nanomedicine in the past decade has given enormous hope for the drug discovery paradigm in the context of many critical diseases, including cancer and neurodegeneration. A fusion of flavonoids with the nano interface has been found to resolve existing pharmacokinetic and toxicity issues by modulating NPs surface chemistry to a great extent. The mechanism through which this change occurs is still not adequately understood and needs to be elaborated further. In-depth perusal into the mechanisms by which flavonoids exhibit their neuroprotective effects and alter properties of nanomaterials to reduce side effects, rendering them nontoxic and multifold beneficial in drug delivery is needed.
Further studies are required to decipher the protein targets and flavonoid–protein interactions in AD to successfully translate the experimental results into clinical outcomes. The central focus should also be given to studies focusing on devising the long-term systemic efficacy, dosage concentrations, and a precise route of administering these drugs for their clinical studies usage before we can consider flavonoid-based nanodrugs in Alzheimer’s treatment. Studies on the combination of flavonoids to enhance the synergistic effects in its correct therapeutic dosage in cells and animal models could also be pursued for a superior outcome. In the context of NPs, more focus should be given to the furtherance of enhancing the synthesis processes and physicochemical properties, including size, shape, structure, surface coating, zeta potential, conjugation procedure, and safety concerns. These parameters are crucial in determining the pharmacokinetics, cellular uptake, and cytotoxicity of nanodrugs and optimizing the delivery systems toward their successful translation into clinics. For example, polymeric NPs show pH and temperature specific enhancement in targeting and efficacy to deliver drugs across physiological barriers.263
These reports altogether suggest that an in-depth understanding of nanomaterials is essential in fabricating appropriate biocompatible nanocarrier corresponding to the targeted site and mechanism of action against AD. The advanced methodologies involved in the enrichment of pharmacological properties of flavonoid nanoformulations with subsequent modifications in their pharmacokinetic and pharmacodynamic profiles need to be deciphered through detailed in vivo studies before we can reach up to some conclusions regarding the future of this amalgamation of natural products and nanomedicine.
Acknowledgments
The authors would like to thank the National Institute of Pharmaceutical Education and Research Hajipur and the Central University of Rajasthan for providing all the required facilities during the preparation of this manuscript. P.P. is receiving a fellowship from the Ministry of Chemicals and Fertilizers, Government of India. We also apologize to many authors whose important works could not be cited due to space limitations.
Glossary
Abbreviations
- 7,8-DHF
7,8-dihydroxyflavone
- ABCA7
ATP-binding cassette transporter A7
- AChE
acetylcholinesterase
- AChEI
acetylcholinesterase Inhibitor
- AD
Alzheimer’s disease
- ADAM10
A disintegrin and metalloprotease 10
- Akt
protein kinase B
- ALS
amyotrophic lateral sclerosis
- AMPK
AMP-activated protein kinase
- APOε4
apolipoprotein E type 4
- APP
amyloid precursor protein
- Aβ
amyloid beta
- BACE
beta-secretase
- BBB
blood–brain barrier
- BChE
butyrylcholinesterase
- BDNF
brain derived neurotropic factor
- BIN1
bridging integrator 1
- Cdk5
cyclin-dependent kinase 5
- CNS
central nervous system
- CLU
clusterin
- CPP
cell-penetrating peptide
- CREB
cAMP response element-binding protein
- EGCG
(−)-epigallocatechin-3-gallate
- Egr1
early growth response protein 1
- ERK
extracellular signal-regulated kinases
- FDA
Food and Drug Administration
- FGF-2
fibroblast growth factor 2
- GFAP
glial fibrillary acidic protein
- GLUT
glucose transporter
- GPx
glutathione peroxidase
- GSH
glutathione
- GSK-3β
glycogen synthase kinase 3β
- HIF-1α
hypoxia-inducible factor-1 alpha
- HO-1
heme oxygenase-1
- IL-1
interleukin-1
- IL-4
interleukin-4
- iNOS
inducible nitric oxide synthase
- IR
insulin receptor
- JNK
c-Jun N-terminal kinase
- LTP
long-term potentiation
- MAPK
mitogen-activated protein kinase
- MDA
malondialdehyde
- MMP9
matrix metallopeptidase 9
- MNPs
metallic nanoparticles
- mTOR
mammalian target of rapamycin
- NFT
neurofibrillary tangles
- NLC
nanostructured lipid carriers
- NMDA
N-methyl-d-aspartate
- NP
nanoparticle
- Nrf2
nuclear factor erythroid 2-related factor 2
- PA2
phospholipase A2
- PARP
poly(ADP-ribose) polymerase
- PBCA
polysorbate 80 coated poly n-butylcyanoacrylate
- PD
Parkinson’s disease
- PEG
polyethylene glycol
- PHD
proline hydroxylase
- PICALM
phosphatidylinositol binding clathrin assembly protein
- PKC
protein kinase C
- PLGA
poly lactic-co-glycolic acid
- PSEN
presenilins
- PI3K
phosphoinositide 3-kinase
- PKA
PPAR-γ, peroxisome proliferator-activated receptor-γ
- ROS
reactive oxygen species
- SAPK
stress-activated protein kinases
- SIRT1
sirtuin1
- SLCP
solid lipid curcumin particle
- SLNs
solid lipid nanoparticles
- SOD
superoxide dismutase
- SORL1
sortilin-related receptor 1
- TNF-α
tumor necrosis factor-α
- TREM2
triggering receptor expressed on myeloid cells 2
- TrkB
tropomyosin-receptor-kinase B
- TSPO
translocator protein
- Unc5c
uncoordinated 5c
The authors declare no competing financial interest.
References
- Giri M.; Zhang M.; Lü Y. (2016) Genes associated with Alzheimer’s disease: an overview and current status. Clin. Interventions Aging 11, 665–681. 10.2147/CIA.S105769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sims R.; Hill M.; Williams J. (2020) The multiplex model of the genetics of Alzheimer’s disease. Nat. Neurosci. 23 (3), 311–322. 10.1038/s41593-020-0599-5. [DOI] [PubMed] [Google Scholar]
- Krance S. H.; Cogo-Moreira H.; Rabin J. S.; Black S. E.; Swardfager W. (2019) Reciprocal Predictive Relationships between Amyloid and Tau Biomarkers in Alzheimer’s Disease Progression: An Empirical Model. J. Neurosci. 39 (37), 7428–7437. 10.1523/JNEUROSCI.1056-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LaFerla F. M.; Green K. N.; Oddo S. (2007) Intracellular amyloid-β in Alzheimer’s disease. Nat. Rev. Neurosci. 8 (7), 499–509. 10.1038/nrn2168. [DOI] [PubMed] [Google Scholar]
- Agostinho P.; Cunha R. A.; Oliveira C. (2010) Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Curr. Pharm. Des. 16 (25), 2766–2778. 10.2174/138161210793176572. [DOI] [PubMed] [Google Scholar]
- Mhatre M.; Floyd R. A.; Hensley K. (2004) Oxidative stress and neuroinflammation in Alzheimer’s disease and amyotrophic lateral sclerosis: common links and potential therapeutic targets. J. Alzheimer's Dis. 6 (2), 147–157. 10.3233/JAD-2004-6206. [DOI] [PubMed] [Google Scholar]
- Crimins J. L.; Pooler A.; Polydoro M.; Luebke J. I.; Spires-Jones T. L. (2013) The intersection of amyloid beta and tau in glutamatergic synaptic dysfunction and collapse in Alzheimer’s disease. Ageing Res. Rev. 12 (3), 757–763. 10.1016/j.arr.2013.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arendt T. (2009) Synaptic degeneration in Alzheimer’s disease. Acta Neuropathol. 118 (1), 167–179. 10.1007/s00401-009-0536-x. [DOI] [PubMed] [Google Scholar]
- Begley D. J. (2004) Delivery of therapeutic agents to the central nervous system: the problems and the possibilities. Pharmacol. Ther. 104 (1), 29–45. 10.1016/j.pharmthera.2004.08.001. [DOI] [PubMed] [Google Scholar]
- Hernando S.; Gartziandia O.; Herran E.; Pedraz J. L.; Igartua M.; Hernandez R. M. (2016) Advances in nanomedicine for the treatment of Alzheimer’s and Parkinson’s diseases. Nanomedicine (London, U. K.) 11 (10), 1267–1285. 10.2217/nnm-2016-0019. [DOI] [PubMed] [Google Scholar]
- Cummings J.; Feldman H. H.; Scheltens P. (2019) The ″rights″ of precision drug development for Alzheimer’s disease. Alzheimer's Res. Ther. 11 (1), 76. 10.1186/s13195-019-0529-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCuistion L. E., Yeager J. J., Winton M. B., and DiMaggio K. V. (2020) Pharmacology-E-Book: A Patient-Centered Nursing Process Approach, Elsevier Health Sciences, . [Google Scholar]
- Roney C.; Kulkarni P.; Arora V.; Antich P.; Bonte F.; Wu A.; Mallikarjuana N.N.; Manohar S.; Liang H.-F.; Kulkarni A. R.; Sung H.-W.; Sairam M.; Aminabhavi T. M. (2005) Targeted nanoparticles for drug delivery through the blood–brain barrier for Alzheimer’s disease. J. Controlled Release 108 (2–3), 193–214. 10.1016/j.jconrel.2005.07.024. [DOI] [PubMed] [Google Scholar]
- Lu C.-T.; Zhao Y.-Z.; Wong H. L.; Cai J.; Peng L.; Tian X.-Q. (2014) Current approaches to enhance CNS delivery of drugs across the brain barriers. Int. J. Nanomed. 9, 2241–2257. 10.2147/IJN.S61288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fonseca-Santos B.; Gremião M. P.; Chorilli M. (2015) Nanotechnology-based drug delivery systems for the treatment of Alzheimer’s disease. Int. J. Nanomed. 10, 4981–5003. 10.2147/IJN.S87148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson B.; Samanta M. K.; Santhi K.; Kumar K. P.; Paramakrishnan N.; Suresh B. (2008) Poly(n-butylcyanoacrylate) nanoparticles coated with polysorbate 80 for the targeted delivery of rivastigmine into the brain to treat Alzheimer’s disease. Brain Res. 1200, 159–168. 10.1016/j.brainres.2008.01.039. [DOI] [PubMed] [Google Scholar]
- Joshi S. A.; Chavhan S. S.; Sawant K. K. (2010) Rivastigmine-loaded PLGA and PBCA nanoparticles: Preparation, optimization, characterization, in vitro and pharmacodynamic studies. Eur. J. Pharm. Biopharm. 76 (2), 189–199. 10.1016/j.ejpb.2010.07.007. [DOI] [PubMed] [Google Scholar]
- Wilson B.; Samanta M. K.; Santhi K.; Kumar K. P.; Paramakrishnan N.; Suresh B. (2008) Targeted delivery of tacrine into the brain with polysorbate 80-coated poly(n-butylcyanoacrylate) nanoparticles. Eur. J. Pharm. Biopharm. 70 (1), 75–84. 10.1016/j.ejpb.2008.03.009. [DOI] [PubMed] [Google Scholar]
- Fazil M.; Md S.; Haque S.; Kumar M.; Baboota S.; Sahni J. K.; Ali J. (2012) Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur. J. Pharm. Sci. 47 (1), 6–15. 10.1016/j.ejps.2012.04.013. [DOI] [PubMed] [Google Scholar]
- Masserini M. (2013) Nanoparticles for brain drug delivery. ISRN Biochem. 2013, 238427–238428. 10.1155/2013/238428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vegh C.; Stokes K.; Ma D.; Wear D.; Cohen J.; Ray S. D.; Pandey S. (2019) A Bird’s-Eye View of the Multiple Biochemical Mechanisms that Propel Pathology of Alzheimer’s Disease: Recent Advances and Mechanistic Perspectives on How to Halt the Disease Progression Targeting Multiple Pathways. J. Alzheimer's Dis. 69, 631–649. 10.3233/JAD-181230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maher P. (2019) The Potential of Flavonoids for the Treatment of Neurodegenerative Diseases. Int. J. Mol. Sci. 20 (12), 3056. 10.3390/ijms20123056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shishtar E.; Rogers G. T.; Blumberg J. B.; Au R.; Jacques P. F. (2020) Long-term dietary flavonoid intake and risk of Alzheimer disease and related dementias in the Framingham Offspring Cohort. Am. J. Clin. Nutr. 112, 343. 10.1093/ajcn/nqaa079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ayaz M.; Sadiq A.; Junaid M.; Ullah F.; Ovais M.; Ullah I.; Ahmed J.; Shahid M. (2019) Flavonoids as Prospective Neuroprotectants and Their Therapeutic Propensity in Aging Associated Neurological Disorders. Front. Aging Neurosci. 11, 154–155. 10.3389/fnagi.2019.00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joshi V.; Mishra R.; Upadhyay A.; Amanullah A.; Poluri K. M.; Singh S.; Kumar A.; Mishra A. (2019) Polyphenolic flavonoid (Myricetin) upregulated proteasomal degradation mechanisms: Eliminates neurodegenerative proteins aggregation. J. Cell. Physiol. 234, 20900–20914. 10.1002/jcp.28695. [DOI] [PubMed] [Google Scholar]
- Amanullah A.; Upadhyay A.; Joshi V.; Mishra R.; Jana N. R.; Mishra A. (2017) Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog. Neurobiol. 159, 1–38. 10.1016/j.pneurobio.2017.08.005. [DOI] [PubMed] [Google Scholar]
- Branquinho Andrade P.; Grosso C.; Valentao P.; Bernardo J. (2016) Flavonoids in neurodegeneration: Limitations and strategies to cross cns barriers [J]. Curr. Med. Chem. 23, 4151–4174. 10.2174/0929867323666160809094934. [DOI] [PubMed] [Google Scholar]
- Es-Safi N.-E.; Ghidouche S.; Ducrot P. H. (2007) Flavonoids: hemisynthesis, reactivity, characterization and free radical scavenging activity. Molecules 12 (9), 2228–2258. 10.3390/12092228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramalho M. J.; Andrade S.; Loureiro J. A.; do Carmo Pereira M. (2020) Nanotechnology to improve the Alzheimer’s disease therapy with natural compounds. Drug Delivery Transl. Res. 10 (2), 380–402. 10.1007/s13346-019-00694-3. [DOI] [PubMed] [Google Scholar]
- DeTure M. A.; Dickson D. W. (2019) The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 14 (1), 32. 10.1186/s13024-019-0333-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melnick V. L., and Dubler N. N. (2012) Alzheimer’s Dementia: Dilemmas in Clinical Research, Springer Science & Business Media. [Google Scholar]
- Bird T. D. (2008) Genetic aspects of Alzheimer disease. Genet. Med. 10 (4), 231–239. 10.1097/GIM.0b013e31816b64dc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alzheimer’s Disease International . World Alzheimer report 2019: attitudes to dementia. In Alzheimer’s Disease International London, UK, 2019. [Google Scholar]
- Prince M. J.World Alzheimer Report 2015: The Global Impact of Dementia: An Analysis of Prevalence, Incidence, Cost and Trends; Alzheimer’s Disease International, 2015. [Google Scholar]
- Prince M.; Ali G. C.; Guerchet M.; Prina A. M.; Albanese E.; Wu Y. T. (2016) Recent global trends in the prevalence and incidence of dementia, and survival with dementia. Alzheimer's Res. Ther. 8 (1), 23. 10.1186/s13195-016-0188-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hussain G.; Zhang L.; Rasul A.; Anwar H.; Sohail M. U.; Razzaq A.; Aziz N.; Shabbir A.; Ali M.; Sun T. (2018) Role of Plant-Derived Flavonoids and Their Mechanism in Attenuation of Alzheimer’s and Parkinson’s Diseases: An Update of Recent Data. Molecules 23 (4), 814. 10.3390/molecules23040814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Awasthi M.; Singh S.; Pandey V. P.; Dwivedi U. N. (2016) Alzheimer’s disease: An overview of amyloid beta dependent pathogenesis and its therapeutic implications along with in silico approaches emphasizing the role of natural products. J. Neurol. Sci. 361, 256–271. 10.1016/j.jns.2016.01.008. [DOI] [PubMed] [Google Scholar]
- Selkoe D. J. (2001) Alzheimer’s disease: genes, proteins, and therapy. Physiol. Rev. 81 (2), 741–766. 10.1152/physrev.2001.81.2.741. [DOI] [PubMed] [Google Scholar]
- Liu P.-P.; Xie Y.; Meng X.-Y.; Kang J.-S. (2019) History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduction and Targeted Therapy 4 (1), 29. 10.1038/s41392-019-0063-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cline E. N.; Bicca M. A.; Viola K. L.; Klein W. L. (2018) The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J. Alzheimer's Dis. 64, S567–S610. 10.3233/JAD-179941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien R. J.; Wong P. C. (2011) Amyloid precursor protein processing and Alzheimer’s disease. Annu. Rev. Neurosci. 34, 185–204. 10.1146/annurev-neuro-061010-113613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hardy J.; Selkoe D. J. (2002) The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297 (5580), 353–356. 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
- Grundke-Iqbal I.; Iqbal K.; Tung Y. C.; Quinlan M.; Wisniewski H. M.; Binder L. I. (1986) Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. U. S. A. 83 (13), 4913–4917. 10.1073/pnas.83.13.4913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blennow K.; Zetterberg H. (2018) Biomarkers for Alzheimer’s disease: current status and prospects for the future. J. Intern. Med. 284 (6), 643–663. 10.1111/joim.12816. [DOI] [PubMed] [Google Scholar]
- Zetterberg H.; Bendlin B. B. (2021) Biomarkers for Alzheimer’s disease—preparing for a new era of disease-modifying therapies. Mol. Psychiatry 26, 296. 10.1038/s41380-020-0721-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Preische O.; Schultz S. A.; Apel A.; Kuhle J.; Kaeser S. A.; Barro C.; Gräber S.; Kuder-Buletta E.; LaFougere C.; Laske C. (2019) Serum neurofilament dynamics predicts neurodegeneration and clinical progression in presymptomatic Alzheimer’s disease. Nature Med. 25 (2), 277–283. 10.1038/s41591-018-0304-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wellington H.; Paterson R. W.; Portelius E.; Törnqvist U.; Magdalinou N.; Fox N. C.; Blennow K.; Schott J. M.; Zetterberg H. (2016) Increased CSF neurogranin concentration is specific to Alzheimer disease. Neurology 86 (9), 829–835. 10.1212/WNL.0000000000002423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molinuevo J. L.; Ayton S.; Batrla R.; Bednar M. M.; Bittner T.; Cummings J.; Fagan A. M.; Hampel H.; Mielke M. M.; Mikulskis A.; O’Bryant S.; Scheltens P.; Sevigny J.; Shaw L. M.; Soares H. D.; Tong G.; Trojanowski J. Q.; Zetterberg H.; Blennow K. (2018) Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 136 (6), 821–853. 10.1007/s00401-018-1932-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parbo P.; Ismail R.; Hansen K. V.; Amidi A.; Marup F. H.; Gottrup H.; Brændgaard H.; Eriksson B. O.; Eskildsen S. F.; Lund T. E.; Tietze A.; Edison P.; Pavese N.; Stokholm M. G.; Borghammer P.; Hinz R.; Aanerud J.; Brooks D. J. (2017) Brain inflammation accompanies amyloid in the majority of mild cognitive impairment cases due to Alzheimer’s disease. Brain 140 (7), 2002–2011. 10.1093/brain/awx120. [DOI] [PubMed] [Google Scholar]
- Luchsinger J. A.; Tang M.-X.; Stern Y.; Shea S.; Mayeux R. (2001) Diabetes Mellitus and Risk of Alzheimer’s Disease and Dementia with Stroke in a Multiethnic Cohort. Am. J. Epidemiol. 154 (7), 635–641. 10.1093/aje/154.7.635. [DOI] [PubMed] [Google Scholar]
- Morris M. S. (2003) Homocysteine and Alzheimer’s disease. Lancet Neurol. 2 (7), 425–428. 10.1016/S1474-4422(03)00438-1. [DOI] [PubMed] [Google Scholar]
- Wyss-Coray T.; Rogers J. (2012) Inflammation in Alzheimer disease-a brief review of the basic science and clinical literature. Cold Spring Harbor Perspect. Med. 2 (1), a006346. 10.1101/cshperspect.a006346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beydoun M. A.; Beydoun H. A.; Wang Y. (2008) Obesity and central obesity as risk factors for incident dementia and its subtypes: a systematic review and meta-analysis. Obes. Rev. 9 (3), 204–218. 10.1111/j.1467-789X.2008.00473.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skoog I.; Gustafson D. (2006) Update on hypertension and Alzheimer’s disease. Neurol. Res. 28 (6), 605–611. 10.1179/016164106X130506. [DOI] [PubMed] [Google Scholar]
- Selkoe D. J. (1996) Amyloid β-protein and the genetics of Alzheimer’s disease. J. Biol. Chem. 271 (31), 18295–18298. 10.1074/jbc.271.31.18295. [DOI] [PubMed] [Google Scholar]
- Wattmo C.; Wallin Å. K. (2017) Early- versus late-onset Alzheimer’s disease in clinical practice: cognitive and global outcomes over 3 years. Alzheimer's Res. Ther. 9 (1), 70. 10.1186/s13195-017-0294-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheuner D.; Eckman C.; Jensen M.; Song X.; Citron M.; Suzuki N.; Bird T.; Hardy J.; Hutton M.; Kukull W. (1996) Secreted amyloid β–protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nature Med. 2 (8), 864–870. 10.1038/nm0896-864. [DOI] [PubMed] [Google Scholar]
- Corder E. H.; Saunders A. M.; Strittmatter W. J.; Schmechel D. E.; Gaskell P. C.; Small G. W.; Roses A. D.; Haines J. L.; Pericak-Vance M. A. (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261 (5123), 921–923. 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
- De Strooper B.; Iwatsubo T.; Wolfe M. S. (2012) Presenilins and γ-secretase: structure, function, and role in Alzheimer Disease. Cold Spring Harbor Perspect. Med. 2 (1), a006304. 10.1101/cshperspect.a006304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiseman F. K.; Al-Janabi T.; Hardy J.; Karmiloff-Smith A.; Nizetic D.; Tybulewicz V. L.; Fisher E. M.; Strydom A. (2015) A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome. Nat. Rev. Neurosci. 16 (9), 564–574. 10.1038/nrn3983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terry A. V.; Buccafusco J. J. (2003) The cholinergic hypothesis of age and Alzheimer’s disease-related cognitive deficits: recent challenges and their implications for novel drug development. J. Pharmacol. Exp. Ther. 306 (3), 821–827. 10.1124/jpet.102.041616. [DOI] [PubMed] [Google Scholar]
- Bartus R. T.; Dean R. L. 3rd; Beer B.; Lippa A. S. (1982) The cholinergic hypothesis of geriatric memory dysfunction. Science 217 (4558), 408–414. 10.1126/science.7046051. [DOI] [PubMed] [Google Scholar]
- Soininen H.; Kosunen O.; Helisalmi S.; Mannermaa A.; Paljarvi L.; Talasniemi S.; Ryynanen M.; Riekkinen P. (1995) A severe loss of choline acetyltransferase in the frontal cortex of Alzheimer patients carrying apolipoprotein epsilon 4 allele. Neurosci. Lett. 187 (2), 79–82. 10.1016/0304-3940(95)11343-6. [DOI] [PubMed] [Google Scholar]
- Schaeffer E. L.; Gattaz W. F. (2008) Cholinergic and glutamatergic alterations beginning at the early stages of Alzheimer disease: participation of the phospholipase A2 enzyme. Psychopharmacology (Berl) 198 (1), 1–27. 10.1007/s00213-008-1092-0. [DOI] [PubMed] [Google Scholar]
- Cai Z.-Y.; Xiao M.; Quazi S. H.; Ke Z.-Y. (2018) Exosomes: a novel therapeutic target for Alzheimer’s disease?. Neural Regener. Res. 13 (5), 930–935. 10.4103/1673-5374.232490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watson L. S.; Hamlett E. D.; Stone T. D.; Sims-Robinson C. (2019) Neuronally derived extracellular vesicles: an emerging tool for understanding Alzheimer’s disease. Mol. Neurodegener. 14 (1), 22. 10.1186/s13024-019-0317-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graykowski D. R.; Wang Y.-Z.; Upadhyay A.; Savas J. N. (2020) The Dichotomous Role of Extracellular Vesicles in the Central Nervous System. iScience 23 (9), 101456. 10.1016/j.isci.2020.101456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin Q.; Ji X.; Lv R.; Pei J.-J.; Du Y.; Shen C.; Hou X. (2020) Targetting Exosomes as a New Biomarker and Therapeutic Approach for Alzheimer’s Disease. Clin. Interventions Aging 15, 195–205. 10.2147/CIA.S240400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown G. C.; Bal-Price A. (2003) Inflammatory neurodegeneration mediated by nitric oxide, glutamate, and mitochondria. Mol. Neurobiol. 27 (3), 325–355. 10.1385/MN:27:3:325. [DOI] [PubMed] [Google Scholar]
- Kinney J. W.; Bemiller S. M.; Murtishaw A. S.; Leisgang A. M.; Salazar A. M.; Lamb B. T. (2018) Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y). 4, 575–590. 10.1016/j.trci.2018.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grammas P. (2011) Neurovascular dysfunction, inflammation and endothelial activation: implications for the pathogenesis of Alzheimer’s disease. J. Neuroinflammation 8, 26. 10.1186/1742-2094-8-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akiyama H.; Barger S.; Barnum S.; Bradt B.; Bauer J.; Cole G. M.; Cooper N. R.; Eikelenboom P.; Emmerling M.; Fiebich B. L.; Finch C. E.; Frautschy S.; Griffin W. S.; Hampel H.; Hull M.; Landreth G.; Lue L.; Mrak R.; Mackenzie I. R.; McGeer P. L.; O’Banion M. K.; Pachter J.; Pasinetti G.; Plata-Salaman C.; Rogers J.; Rydel R.; Shen Y.; Streit W.; Strohmeyer R.; Tooyoma I.; Van Muiswinkel F. L.; Veerhuis R.; Walker D.; Webster S.; Wegrzyniak B.; Wenk G.; Wyss-Coray T. (2000) Inflammation and Alzheimer’s disease. Neurobiol. Aging 21 (3), 383–421. 10.1016/S0197-4580(00)00124-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Combs C. K.; Johnson D. E.; Karlo J. C.; Cannady S. B.; Landreth G. E. (2000) Inflammatory mechanisms in Alzheimer’s disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists. J. Neurosci. 20 (2), 558–567. 10.1523/JNEUROSCI.20-02-00558.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitazawa M.; Yamasaki T. R.; LaFerla F. M. (2004) Microglia as a potential bridge between the amyloid beta-peptide and tau. Ann. N. Y. Acad. Sci. 1035, 85–103. 10.1196/annals.1332.006. [DOI] [PubMed] [Google Scholar]
- Akama K. T.; Van Eldik L. J. (2000) Beta-amyloid stimulation of inducible nitric-oxide synthase in astrocytes is interleukin-1beta- and tumor necrosis factor-alpha (TNFalpha)-dependent, and involves a TNFalpha receptor-associated factor- and NFkappaB-inducing kinase-dependent signaling mechanism. J. Biol. Chem. 275 (11), 7918–7924. 10.1074/jbc.275.11.7918. [DOI] [PubMed] [Google Scholar]
- Blasko I.; Stampfer-Kountchev M.; Robatscher P.; Veerhuis R.; Eikelenboom P.; Grubeck-Loebenstein B. (2004) How chronic inflammation can affect the brain and support the development of Alzheimer’s disease in old age: the role of microglia and astrocytes. Aging Cell 3 (4), 169–176. 10.1111/j.1474-9728.2004.00101.x. [DOI] [PubMed] [Google Scholar]
- Johnstone M.; Gearing A. J.; Miller K. M. (1999) A central role for astrocytes in the inflammatory response to beta-amyloid; chemokines, cytokines and reactive oxygen species are produced. J. Neuroimmunol. 93 (1–2), 182–193. 10.1016/S0165-5728(98)00226-4. [DOI] [PubMed] [Google Scholar]
- Retz W., Gsell W., Münch G., Rösler M., and Riederer P. (1998) Free radicals in Alzheimer’s disease. In Alzheimer’s Disease—From Basic Research to Clinical Applications, pp 221–236, Springer. [DOI] [PubMed] [Google Scholar]
- Fahnestock M.; Garzon D.; Holsinger R. M.; Michalski B. (2002) Neurotrophic factors and Alzheimer’s disease: are we focusing on the wrong molecule?. J. Neural Transm., Suppl. 62 (62), 241–252. 10.1007/978-3-7091-6139-5_22. [DOI] [PubMed] [Google Scholar]
- Schindowski K.; Belarbi K.; Buée L. (2008) Neurotrophic factors in Alzheimer’s disease: role of axonal transport. Genes, Brain Behav. 7 (Suppl 1 (1)), 43–56. 10.1111/j.1601-183X.2007.00378.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen X. Q.; Sawa M.; Mobley W. C. (2018) Dysregulation of neurotrophin signaling in the pathogenesis of Alzheimer disease and of Alzheimer disease in Down syndrome. Free Radical Biol. Med. 114, 52–61. 10.1016/j.freeradbiomed.2017.10.341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Awasthi M.; Upadhyay A. K.; Singh S.; Pandey V. P.; Dwivedi U. N. (2018) Terpenoids as promising therapeutic molecules against Alzheimer’s disease: amyloid beta- and acetylcholinesterase-directed pharmacokinetic and molecular docking analyses. Mol. Simul. 44 (1), 1–11. 10.1080/08927022.2017.1334880. [DOI] [Google Scholar]
- Upadhyay A.; Mishra A. (2018) Amyloids of multiple species: are they helpful in survival?. Biological Reviews 93 (3), 1363. 10.1111/brv.12399. [DOI] [PubMed] [Google Scholar]
- Ostrowski S. M.; Wilkinson B. L.; Golde T. E.; Landreth G. (2007) Statins reduce amyloid-beta production through inhibition of protein isoprenylation. J. Biol. Chem. 282 (37), 26832–26844. 10.1074/jbc.M702640200. [DOI] [PubMed] [Google Scholar]
- Eckert G. P.; Müller W. E.; Wood G. W. (2007) Cholesterol-lowering drugs and Alzheimer’s disease. Future Lipidol. 2 (4), 423–432. 10.2217/17460875.2.4.423. [DOI] [Google Scholar]
- Rich J. B.; Rasmusson D.; Folstein M. F.; Carson K. A.; Kawas C.; Brandt J. (1995) Nonsteroidal anti-inflammatory drugs in Alzheimer’s disease. Neurology 45 (1), 51–55. 10.1212/WNL.45.1.51. [DOI] [PubMed] [Google Scholar]
- Anekonda T. S.; Reddy P. H. (2005) Can herbs provide a new generation of drugs for treating Alzheimer’s disease?. Brain Res. Rev. 50 (2), 361–376. 10.1016/j.brainresrev.2005.09.001. [DOI] [PubMed] [Google Scholar]
- Hock C.; Konietzko U.; Streffer J. R; Tracy J.; Signorell A.; Muller-Tillmanns B.; Lemke U.; Henke K.; Moritz E.; Garcia E.; Wollmer M.A.; Umbricht D.; de Quervain D. J.F; Hofmann M.; Maddalena A.; Papassotiropoulos A.; Nitsch R. M (2003) Antibodies against β-amyloid slow cognitive decline in Alzheimer’s disease. Neuron 38 (4), 547–554. 10.1016/S0896-6273(03)00294-0. [DOI] [PubMed] [Google Scholar]
- Ferreira-Vieira T. H.; Guimaraes I. M.; Silva F. R.; Ribeiro F. M. (2016) Alzheimer’s disease: Targeting the Cholinergic System. Curr. Neuropharmacol. 14 (1), 101–115. 10.2174/1570159X13666150716165726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kabir M. T.; Uddin M. S.; Begum M. M.; Thangapandiyan S.; Rahman M. S.; Aleya L.; Mathew B.; Ahmed M.; Barreto G. E.; Ashraf G. M. (2019) Cholinesterase Inhibitors for Alzheimer’s Disease: Multitargeting Strategy Based on Anti-Alzheimer’s Drugs Repositioning. Curr. Pharm. Des. 25 (33), 3519–3535. 10.2174/1381612825666191008103141. [DOI] [PubMed] [Google Scholar]
- Anand P.; Singh B. (2013) A review on cholinesterase inhibitors for Alzheimer’s disease. Arch. Pharmacal Res. 36 (4), 375–399. 10.1007/s12272-013-0036-3. [DOI] [PubMed] [Google Scholar]
- Rosini M.; Simoni E.; Bartolini M.; Cavalli A.; Ceccarini L.; Pascu N.; McClymont D. W.; Tarozzi A.; Bolognesi M. L.; Minarini A.; Tumiatti V.; Andrisano V.; Mellor I. R.; Melchiorre C. (2008) Inhibition of acetylcholinesterase, β-amyloid aggregation, and NMDA receptors in Alzheimer’s disease: a promising direction for the multi-target-directed ligands gold rush. J. Med. Chem. 51 (15), 4381–4384. 10.1021/jm800577j. [DOI] [PubMed] [Google Scholar]
- Birks J. S., Chong L. Y., and Grimley Evans J. (2015) Rivastigmine for Alzheimer’s disease. Cochrane Database of Systematic Reviews, 10.1002/14651858.CD001191.pub4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Unzeta M., Esteban G., Bolea I., Fogel W. A., Ramsay R. R., Youdim M. B. H., Tipton K. F., and Marco-Contelles J. (2016) Multi-Target Directed Donepezil-Like Ligands for Alzheimer’s Disease. Front. Neurosci. 10, (205), , 10.3389/fnins.2016.00205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomsen T.; Kewitz H. (1990) Selective inhibition of human acetylcholinesterase by galanthamine in vitro and in vivo. Life Sci. 46 (21), 1553–1558. 10.1016/0024-3205(90)90429-U. [DOI] [PubMed] [Google Scholar]
- Woodruff-Pak D. S.; Vogel R. W.; Wenk G. L. (2001) Galantamine: effect on nicotinic receptor binding, acetylcholinesterase inhibition, and learning. Proc. Natl. Acad. Sci. U. S. A. 98 (4), 2089–2094. 10.1073/pnas.98.4.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agatonovic-Kustrin S.; Kettle C.; Morton D. W. (2018) A molecular approach in drug development for Alzheimer’s disease. Biomed. Pharmacother. 106, 553–565. 10.1016/j.biopha.2018.06.147. [DOI] [PubMed] [Google Scholar]
- Cummings J. L.; Tong G.; Ballard C. (2019) Treatment Combinations for Alzheimer’s Disease: Current and Future Pharmacotherapy Options. J. Alzheimer's Dis. 67 (3), 779–794. 10.3233/JAD-180766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tiwari S.; Atluri V.; Kaushik A.; Yndart A.; Nair M. (2019) Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. Int. J. Nanomed. 14, 5541–5554. 10.2147/IJN.S200490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karthivashan G.; Ganesan P.; Park S.-Y.; Kim J.-S.; Choi D.-K. (2018) Therapeutic strategies and nano-drug delivery applications in management of ageing Alzheimer’s disease. Drug Delivery 25 (1), 307–320. 10.1080/10717544.2018.1428243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mullard A. (2017) BACE inhibitor bust in Alzheimer trial. Nat. Rev. Drug Discovery 16 (3), 154–155. [DOI] [PubMed] [Google Scholar]
- Alva G.; Cummings J. L. (2008) Relative tolerability of Alzheimer’s disease treatments. Psychiatry (Edgmont) 5 (11), 27–36. [PMC free article] [PubMed] [Google Scholar]
- Dhillon S. (2011) Rivastigmine transdermal patch: a review of its use in the management of dementia of the Alzheimer’s type. Drugs 71 (9), 1209–1231. 10.2165/11206380-000000000-00000. [DOI] [PubMed] [Google Scholar]
- Cummings J.; Lefèvre G.; Small G.; Appel-Dingemanse S. (2007) Pharmacokinetic rationale for the rivastigmine patch. Neurology 69 (4 Suppl 1), S10–S13. 10.1212/01.wnl.0000281846.40390.50. [DOI] [PubMed] [Google Scholar]
- Upadhyay R. K. (2014) Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res. Int. 2014, 869268–869269. 10.1155/2014/869269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong X. (2018) Current strategies for brain drug delivery. Theranostics 8 (6), 1481. 10.7150/thno.21254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilroy K. D.; Ruditskiy A.; Peng H.-C.; Qin D.; Xia Y. (2016) Bimetallic nanocrystals: syntheses, properties, and applications. Chem. Rev. 116 (18), 10414–10472. 10.1021/acs.chemrev.6b00211. [DOI] [PubMed] [Google Scholar]
- Huang C.; Chen X.; Xue Z.; Wang T. (2020) Effect of structure: A new insight into nanoparticle assemblies from inanimate to animate. Science advances 6 (20), eaba1321 10.1126/sciadv.aba1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeevanandam J.; Barhoum A.; Chan Y. S.; Dufresne A.; Danquah M. K. (2018) Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J. Nanotechnol. 9, 1050–1074. 10.3762/bjnano.9.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elechiguerra J. L.; Larios-Lopez L.; Liu C.; Garcia-Gutierrez D.; Camacho-Bragado A.; Yacaman M. J. (2005) Corrosion at the Nanoscale: The Case of Silver Nanowires and Nanoparticles. Chem. Mater. 17 (24), 6042–6052. 10.1021/cm051532n. [DOI] [Google Scholar]
- Ahamed M.; Alsalhi M. S.; Siddiqui M. K. (2010) Silver nanoparticle applications and human health. Clin. Chim. Acta 411 (23–24), 1841–1848. 10.1016/j.cca.2010.08.016. [DOI] [PubMed] [Google Scholar]
- Prasad M.; Lambe U. P.; Brar B.; Shah I.; J MManimegalai.; Ranjan K.; Rao R.; Kumar S.; Mahant S.; Khurana S. K.; Iqbal H. M.N.; Dhama K.; Misri J.; Prasad G. (2018) Nanotherapeutics: An insight into healthcare and multi-dimensional applications in medical sector of the modern world. Biomed. Pharmacother. 97, 1521–1537. 10.1016/j.biopha.2017.11.026. [DOI] [PubMed] [Google Scholar]
- Patra J. K.; Das G.; Fraceto L. F.; Campos E. V. R.; Rodriguez-Torres M. D. P.; Acosta-Torres L. S.; Diaz-Torres L. A.; Grillo R.; Swamy M. K.; Sharma S.; Habtemariam S.; Shin H.-S. (2018) Nano based drug delivery systems: recent developments and future prospects. J. Nanobiotechnol. 16 (1), 70–71. 10.1186/s12951-018-0392-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeevanandam J.; Chan Y. S.; Danquah M. K. (2016) Nano-formulations of drugs: Recent developments, impact and challenges. Biochimie 128–129, 99–112. 10.1016/j.biochi.2016.07.008. [DOI] [PubMed] [Google Scholar]
- Sherine J.; Upadhyay A.; Mishra A.; Kumar D.; Pal S.; Harinipriya S. (2019) Ag(I) and Au(III) Mercaptobenzothiazole complexes induced apoptotic cell death. Sci. Rep. 9 (1), 621. 10.1038/s41598-018-36801-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaushik A.; Jayant R. D.; Bhardwaj V.; Nair M. (2018) Personalized nanomedicine for CNS diseases. Drug Discovery Today 23 (5), 1007–1015. 10.1016/j.drudis.2017.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pajouhesh H.; Lenz G. R. (2005) Medicinal chemical properties of successful central nervous system drugs. NeuroRx 2 (4), 541–553. 10.1602/neurorx.2.4.541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phachonpai W.; Wattanathorn J.; Muchimapura S.; Tong-Un T.; Preechagoon D. (2010) Neuroprotective effect of quercetin encapsulated liposomes: a novel therapeutic strategy against Alzheimer’s disease. Am. J. Appl. Sci. 7 (4), 480–485. 10.3844/ajassp.2010.480.485. [DOI] [Google Scholar]
- Wilson B. (2009) Brain targeting PBCA nanoparticles and the blood–brain barrier. Nanomedicine 4 (5), 499–502. 10.2217/nnm.09.29. [DOI] [PubMed] [Google Scholar]
- Hanafy A. S.; Farid R. M.; ElGamal S. S. (2015) Complexation as an approach to entrap cationic drugs into cationic nanoparticles administered intranasally for Alzheimer’s disease management: preparation and detection in rat brain. Drug Dev. Ind. Pharm. 41 (12), 2055–2068. 10.3109/03639045.2015.1062897. [DOI] [PubMed] [Google Scholar]
- Luppi B.; Bigucci F.; Corace G.; Delucca A.; Cerchiara T.; Sorrenti M.; Catenacci L.; Di Pietra A. M.; Zecchi V. (2011) Albumin nanoparticles carrying cyclodextrins for nasal delivery of the anti-Alzheimer drug tacrine. Eur. J. Pharm. Sci. 44 (4), 559–565. 10.1016/j.ejps.2011.10.002. [DOI] [PubMed] [Google Scholar]
- Liu Z.; Gao X.; Kang T.; Jiang M.; Miao D.; Gu G.; Hu Q.; Song Q.; Yao L.; Tu Y.; Chen H.; Jiang X.; Chen J. (2013) B6 peptide-modified PEG-PLA nanoparticles for enhanced brain delivery of neuroprotective peptide. Bioconjugate Chem. 24 (6), 997–1007. 10.1021/bc400055h. [DOI] [PubMed] [Google Scholar]
- Nagpal K.; Singh S. K.; Mishra D. N. (2013) Optimization of brain targeted chitosan nanoparticles of Rivastigmine for improved efficacy and safety. Int. J. Biol. Macromol. 59, 72–83. 10.1016/j.ijbiomac.2013.04.024. [DOI] [PubMed] [Google Scholar]
- Pathak Y. V.Surface Modification of Nanoparticles for Targeted Drug Delivery; Springer, 2019. [Google Scholar]
- Khan S.; Baboota S.; Ali J.; Khan S.; Narang R. S.; Narang J. K. (2015) Nanostructured lipid carriers: An emerging platform for improving oral bioavailability of lipophilic drugs. Int. J. Pharm. Investig 5 (4), 182–191. 10.4103/2230-973X.167661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhuang C. Y.; Li N.; Wang M.; Zhang X. N.; Pan W. S.; Peng J. J.; Pan Y. S.; Tang X. (2010) Preparation and characterization of vinpocetine loaded nanostructured lipid carriers (NLC) for improved oral bioavailability. Int. J. Pharm. 394 (1–2), 179–185. 10.1016/j.ijpharm.2010.05.005. [DOI] [PubMed] [Google Scholar]
- Illum L. (2000) Transport of drugs from the nasal cavity to the central nervous system. Eur. J. Pharm. Sci. 11 (1), 1–18. 10.1016/S0928-0987(00)00087-7. [DOI] [PubMed] [Google Scholar]
- Sonvico F.; Clementino A.; Buttini F.; Colombo G.; Pescina S.; Stanisçuaski Guterres S.; Raffin Pohlmann A.; Nicoli S. (2018) Surface-Modified Nanocarriers for Nose-to-Brain Delivery: From Bioadhesion to Targeting. Pharmaceutics 10 (1), 34. 10.3390/pharmaceutics10010034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Z.-Z.; Zhang Y.-Q.; Wang Z.-Z.; Wu K.; Lou J.-N.; Qi X.-R. (2013) Enhanced brain distribution and pharmacodynamics of rivastigmine by liposomes following intranasal administration. Int. J. Pharm. 452 (1), 344–354. 10.1016/j.ijpharm.2013.05.009. [DOI] [PubMed] [Google Scholar]
- Al Asmari A. K., Ullah Z., Tariq M., and Fatani A. (2016) Preparation, characterization, and in vivo evaluation of intranasally administered liposomal formulation of donepezil, in Drug Design, Development and Therapy (pp 205–215). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corace G.; Angeloni C.; Malaguti M.; Hrelia S.; Stein P. C.; Brandl M.; Gotti R.; Luppi B. (2014) Multifunctional liposomes for nasal delivery of the anti-Alzheimer drug tacrine hydrochloride. J. Liposome Res. 24 (4), 323–335. 10.3109/08982104.2014.899369. [DOI] [PubMed] [Google Scholar]
- Mufamadi M. S.; Choonara Y. E.; Kumar P.; Modi G.; Naidoo D.; van Vuuren S.; Ndesendo V. M. K.; Toit LCd; Iyuke S. E.; Pillay V. (2013) Ligand-functionalized nanoliposomes for targeted delivery of galantamine. Int. J. Pharm. 448 (1), 267–281. 10.1016/j.ijpharm.2013.03.037. [DOI] [PubMed] [Google Scholar]
- Ismail M. F.; Elmeshad A. N.; Salem NA-H (2013) Potential therapeutic effect of nanobased formulation of rivastigmine on rat model of Alzheimer’s disease. Int. J. Nanomed. 8, 393–406. 10.2147/IJN.S39232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onoue S.; Yamada S.; Chan H. K. (2014) Nanodrugs: pharmacokinetics and safety. Int. J. Nanomed. 9, 1025–1037. 10.2147/IJN.S38378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Jong W. H.; Borm P. J. A. (2008) Drug delivery and nanoparticles:applications and hazards. Int. J. Nanomed. 3 (2), 133–149. 10.2147/IJN.S596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cummings J.; Lee G.; Ritter A.; Sabbagh M.; Zhong K. (2019) Alzheimer’s disease drug development pipeline: 2019. Alzheimer’s & dementia (New York, N Y) 5, 272–293. 10.1016/j.trci.2019.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doody R. S.; Raman R.; Farlow M.; Iwatsubo T.; Vellas B.; Joffe S.; Kieburtz K.; He F.; Sun X.; Thomas R. G.; Aisen P. S.; Siemers E.; Sethuraman G.; Mohs R. (2013) A phase 3 trial of semagacestat for treatment of Alzheimer’s disease. N. Engl. J. Med. 369 (4), 341–350. 10.1056/NEJMoa1210951. [DOI] [PubMed] [Google Scholar]
- Henley D.; Raghavan N.; Sperling R.; Aisen P.; Raman R.; Romano G. (2019) Preliminary Results of a Trial of Atabecestat in Preclinical Alzheimer’s Disease. N. Engl. J. Med. 380 (15), 1483–1485. 10.1056/NEJMc1813435. [DOI] [PubMed] [Google Scholar]
- Vandenberghe R.; Rinne J. O.; Boada M.; Katayama S.; Scheltens P.; Vellas B.; Tuchman M.; Gass A.; Fiebach J. B.; Hill D.; Lobello K.; Li D.; McRae T.; Lucas P.; Evans I.; Booth K.; Luscan G.; Wyman B. T.; Hua L.; Yang L.; Brashear H. R.; Black R. S. (2016) Bapineuzumab for mild to moderate Alzheimer’s disease in two global, randomized, phase 3 trials. Alzheimer's Res. Ther. 8 (1), 18. 10.1186/s13195-016-0189-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badhwar R., and Upadhyay A. (2020) Modulation of Cellular Protein Quality Control Pathways Using Small Natural Molecules. In Bioactive Natural products in Drug Discovery. Springer Singapore, Singapore, Singh J., Meshram V., and Gupta M., Eds.) pp 609–641. [Google Scholar]
- Kumar S., and Pandey A. K.. Chemistry and biological activities of flavonoids: an overview. Sci. World J. 2013;2013.1. 10.1155/2013/162750 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Panche A. N.; Diwan A. D.; Chandra S. R. (2016) Flavonoids: an overview. J. Nutr. Sci. 5, e46–e47. 10.1017/jns.2016.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berhow M. A.Modern Analytical Techniques for Flavonoid Determination. In: Buslig B. S., and Manthey J. A., Eds. (2002) Flavonoids in Cell Function, (pp 61–76), Springer US, Boston, MA. [DOI] [PubMed] [Google Scholar]
- Williams C. A.; Grayer R. J. (2004) Anthocyanins and other flavonoids. Nat. Prod. Rep. 21 (4), 539–573. 10.1039/b311404j. [DOI] [PubMed] [Google Scholar]
- Iriti M., and Varoni E. M. (2015) Brief introduction to polyphenols, bioactive phytochemicals for human health. Recent Advances in Medicinal Chemistry, Vol. 2, issue (3), . [Google Scholar]
- Corradini E.; Foglia P.; Giansanti P.; Gubbiotti R.; Samperi R.; Lagana A. (2011) Flavonoids: chemical properties and analytical methodologies of identification and quantitation in foods and plants. Nat. Prod. Res. 25 (5), 469–495. 10.1080/14786419.2010.482054. [DOI] [PubMed] [Google Scholar]
- Gropper S. S., and Smith J. L. (2012) Advanced Nutrition and Human Metabolism, Cengage Learning. [Google Scholar]
- Khan E.; Tawani A.; Mishra S. K.; Verma A. K.; Upadhyay A.; Kumar M.; Sandhir R.; Mishra A.; Kumar A. (2018) Myricetin reduces toxic level of CAG repeats RNA in Huntington’s Disease (HD) and Spino Cerebellar Ataxia (SCAs). ACS Chem. Biol. 13, 180. 10.1021/acschembio.7b00699. [DOI] [PubMed] [Google Scholar]
- Varshney R.; Mishra R.; Das N.; Sircar D.; Roy P. (2019) A comparative analysis of various flavonoids in the regulation of obesity and diabetes: An in vitro and in vivo study. J. Funct. Foods 59, 194–205. 10.1016/j.jff.2019.05.004. [DOI] [Google Scholar]
- Varshney R.; Gupta S.; Roy P. (2017) Cytoprotective effect of kaempferol against palmitic acid-induced pancreatic β-cell death through modulation of autophagy via AMPK/mTOR signaling pathway. Mol. Cell. Endocrinol. 448, 1–20. 10.1016/j.mce.2017.02.033. [DOI] [PubMed] [Google Scholar]
- Braak H., de Vos R. A., Jansen E. N., Bratzke H., and Braak E. (1998) Neuropathological hallmarks of Alzheimer’s and Parkinson’s diseases. In Progress in Brain Research, pp 267–285, Elsevier. [DOI] [PubMed] [Google Scholar]
- Lue L.-F.; Brachova L.; Civin W. H.; Rogers J. (1996) Inflammation, Aβ deposition, and neurofibrillary tangle formation as correlates of Alzheimer’s disease neurodegeneration. J. Neuropathol. Exp. Neurol. 55 (10), 1083–1088. 10.1097/00005072-199655100-00008. [DOI] [PubMed] [Google Scholar]
- Hemanth Kumar B.; Dinesh Kumar B.; Diwan P. V. (2017) Hesperidin, a citrus flavonoid, protects against l-methionine-induced hyperhomocysteinemia by abrogation of oxidative stress, endothelial dysfunction and neurotoxicity in Wistar rats. Pharm. Biol. 55 (1), 146–155. 10.1080/13880209.2016.1231695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee S.; Suh S.; Kim S. (2000) Protective effects of the green tea polyphenol (−)-epigallocatechin gallate against hippocampal neuronal damage after transient global ischemia in gerbils. Neurosci. Lett. 287 (3), 191–194. 10.1016/S0304-3940(00)01159-9. [DOI] [PubMed] [Google Scholar]
- Levites Y.; Weinreb O.; Maor G.; Youdim M. B.; Mandel S. (2001) Green tea polyphenol (−)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neurochem. 78 (5), 1073–1082. 10.1046/j.1471-4159.2001.00490.x. [DOI] [PubMed] [Google Scholar]
- Xu Z.; Chen S.; Li X.; Luo G.; Li L.; Le W. (2006) Neuroprotective effects of (−)-epigallocatechin-3-gallate in a transgenic mouse model of amyotrophic lateral sclerosis. Neurochem. Res. 31 (10), 1263–1269. 10.1007/s11064-006-9166-z. [DOI] [PubMed] [Google Scholar]
- Park S.-S.; Bae I.; Lee Y. J. (2008) Flavonoids-induced accumulation of hypoxia-inducible factor (HIF)-1α/2α is mediated through chelation of iron. J. Cell. Biochem. 103 (6), 1989–1998. 10.1002/jcb.21588. [DOI] [PubMed] [Google Scholar]
- Lou H.; Jing X.; Wei X.; Shi H.; Ren D.; Zhang X. (2014) Naringenin protects against 6-OHDA-induced neurotoxicity via activation of the Nrf2/ARE signaling pathway. Neuropharmacology 79, 380–388. 10.1016/j.neuropharm.2013.11.026. [DOI] [PubMed] [Google Scholar]
- Jing X.; Shi H.; Zhu X.; Wei X.; Ren M.; Han M.; Ren D.; Lou H. (2015) Eriodictyol Attenuates β-Amyloid 25–35 Peptide-Induced Oxidative Cell Death in Primary Cultured Neurons by Activation of Nrf2. Neurochem. Res. 40 (7), 1463–1471. 10.1007/s11064-015-1616-z. [DOI] [PubMed] [Google Scholar]
- de Oliveira M. R.; da Costa Ferreira G.; Brasil F. B.; Peres A. (2018) Pinocembrin suppresses H 2 O 2-induced mitochondrial dysfunction by a mechanism dependent on the Nrf2/HO-1 axis in SH-SY5Y cells. Mol. Neurobiol. 55 (2), 989–1003. 10.1007/s12035-016-0380-7. [DOI] [PubMed] [Google Scholar]
- Upadhyay A. (2021) Natural compounds in the regulation of proteostatic pathways: An invincible artillery against stress, ageing, and diseases. Acta Pharm. Sin. B. 10.1016/j.apsb.2021.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee H.; Kim Y. O.; Kim H.; Kim S. Y.; Noh H. S.; Kang S. S.; Cho G. J.; Choi W. S.; Suk K. (2003) Flavonoid wogonin from medicinal herb is neuroprotective by inhibiting inflammatory activation of microglia. FASEB J. 17 (13), 1943–1944. 10.1096/fj.03-0057fje. [DOI] [PubMed] [Google Scholar]
- Wang X.; Chen S.; Ma G.; Ye M.; Lu G. (2005) Genistein protects dopaminergic neurons by inhibiting microglial activation. NeuroReport 16 (3), 267–270. 10.1097/00001756-200502280-00013. [DOI] [PubMed] [Google Scholar]
- Li R.; Huang Y. G.; Fang D.; Le W. D. (2004) (−)-Epigallocatechin gallate inhibits lipopolysaccharide-induced microglial activation and protects against inflammation-mediated dopaminergic neuronal injury. J. Neurosci. Res. 78 (5), 723–731. 10.1002/jnr.20315. [DOI] [PubMed] [Google Scholar]
- Chen J.-C.; Ho F.-M.; Pei-Dawn Lee Chao; Chen C.-P.; Jeng K.-C. G.; Hsu H.-B.; Lee S.-T.; Wen Tung Wu; Lin W.-W. (2005) Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IkappaB kinase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur. J. Pharmacol. 521 (1–3), 9–20. 10.1016/j.ejphar.2005.08.005. [DOI] [PubMed] [Google Scholar]
- Zheng L. T.; Ock J.; Kwon B. M.; Suk K. (2008) Suppressive effects of flavonoid fisetin on lipopolysaccharide-induced microglial activation and neurotoxicity. Int. Immunopharmacol. 8 (3), 484–494. 10.1016/j.intimp.2007.12.012. [DOI] [PubMed] [Google Scholar]
- Kim H. P.; Son K. H.; Chang H. W.; Kang S. S. (2004) Anti-inflammatory plant flavonoids and cellular action mechanisms. J. Pharmacol. Sci. 96, 229–245. 10.1254/jphs.CRJ04003X. [DOI] [PubMed] [Google Scholar]
- Spencer J. P. (2008) Flavonoids: modulators of brain function?. Br. J. Nutr. 99 (E Suppl 1), ES60–ES77. 10.1017/S0007114508965776. [DOI] [PubMed] [Google Scholar]
- Lee H. J.; Noh Y. H.; Lee D. Y.; Kim Y. S.; Kim K. Y.; Chung Y. H.; Lee W. B.; Kim S. S. (2005) Baicalein attenuates 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y cells. Eur. J. Cell Biol. 84 (11), 897–905. 10.1016/j.ejcb.2005.07.003. [DOI] [PubMed] [Google Scholar]
- Ishikawa Y.; Kitamura M. (2000) Anti-apoptotic effect of quercetin: intervention in the JNK- and ERK-mediated apoptotic pathways. Kidney Int. 58 (3), 1078–1087. 10.1046/j.1523-1755.2000.00265.x. [DOI] [PubMed] [Google Scholar]
- Schroeter H.; Spencer J. P.; Rice-Evans C.; Williams R. J. (2001) Flavonoids protect neurons from oxidized low-density-lipoprotein-induced apoptosis involving c-Jun N-terminal kinase (JNK), c-Jun and caspase-3. Biochem. J. 358 (3), 547–557. 10.1042/bj3580547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sachdeva A. K.; Kuhad A.; Chopra K. (2014) Naringin ameliorates memory deficits in experimental paradigm of Alzheimer’s disease by attenuating mitochondrial dysfunction. Pharmacol., Biochem. Behav. 127, 101–110. 10.1016/j.pbb.2014.11.002. [DOI] [PubMed] [Google Scholar]
- Gu X. H.; Xu L. J.; Liu Z. Q.; Wei B.; Yang Y. J.; Xu G. G.; Yin X. P.; Wang W. (2016) The flavonoid baicalein rescues synaptic plasticity and memory deficits in a mouse model of Alzheimer’s disease. Behav. Brain Res. 311, 309–321. 10.1016/j.bbr.2016.05.052. [DOI] [PubMed] [Google Scholar]
- Han Q.; Wang X.; Cai S.; Liu X.; Zhang Y.; Yang L.; Wang C.; Yang R. (2018) Quercetin nanoparticles with enhanced bioavailability as multifunctional agents toward amyloid induced neurotoxicity. J. Mater. Chem. B 6 (9), 1387–1393. 10.1039/C7TB03053C. [DOI] [PubMed] [Google Scholar]
- Youdim K. A.; Joseph J. A. (2001) A possible emerging role of phytochemicals in improving age-related neurological dysfunctions: a multiplicity of effects. Free Radical Biol. Med. 30 (6), 583–594. 10.1016/S0891-5849(00)00510-4. [DOI] [PubMed] [Google Scholar]
- Williams R. J.; Spencer J. P. (2012) Flavonoids, cognition, and dementia: actions, mechanisms, and potential therapeutic utility for Alzheimer disease. Free Radical Biol. Med. 52 (1), 35–45. 10.1016/j.freeradbiomed.2011.09.010. [DOI] [PubMed] [Google Scholar]
- Flanagan E.; Müller M.; Hornberger M.; Vauzour D. (2018) Impact of Flavonoids on Cellular and Molecular Mechanisms Underlying Age-Related Cognitive Decline and Neurodegeneration. Curr. Nutr. Rep. 7 (2), 49–57. 10.1007/s13668-018-0226-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maher P.; Akaishi T.; Abe K. (2006) Flavonoid fisetin promotes ERK-dependent long-term potentiation and enhances memory. Proc. Natl. Acad. Sci. U. S. A. 103 (44), 16568–16573. 10.1073/pnas.0607822103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu B.; Li X. X.; He G. R.; Hu J. J.; Mu X.; Tian S.; Du G. H. (2010) Luteolin promotes long-term potentiation and improves cognitive functions in chronic cerebral hypoperfused rats. Eur. J. Pharmacol. 627 (1–3), 99–105. 10.1016/j.ejphar.2009.10.038. [DOI] [PubMed] [Google Scholar]
- Guzzi C.; Colombo L.; Luigi A.; Salmona M.; Nicotra F.; Airoldi C. (2017) Flavonoids and Their Glycosides as Anti-amyloidogenic Compounds: Aβ1–42 Interaction Studies to Gain New Insights into Their Potential for Alzheimer’s Disease Prevention and Therapy. Chem. - Asian J. 12 (1), 67–75. 10.1002/asia.201601291. [DOI] [PubMed] [Google Scholar]
- Uddin M. S.; Mamun A. A.; Hossain M. S.; Akter F.; Iqbal M. A.; Asaduzzaman M. (2016) Exploring the Effect of Phyllanthus emblica L. on Cognitive Performance, Brain Antioxidant Markers and Acetylcholinesterase Activity in Rats: Promising Natural Gift for the Mitigation of Alzheimer’s Disease. Ann. Neurosci. 23 (4), 218–229. 10.1159/000449482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ono K.; Yoshiike Y.; Takashima A.; Hasegawa K.; Naiki H.; Yamada M. (2003) Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: implications for the prevention and therapeutics of Alzheimer’s disease. J. Neurochem. 87 (1), 172–181. 10.1046/j.1471-4159.2003.01976.x. [DOI] [PubMed] [Google Scholar]
- Ali M. Y.; Jannat S.; Edraki N.; Das S.; Chang W. K.; Kim H. C.; Park S. K.; Chang M. S. (2019) Flavanone glycosides inhibit β-site amyloid precursor protein cleaving enzyme 1 and cholinesterase and reduce Aβ aggregation in the amyloidogenic pathway. Chem.-Biol. Interact. 309, 108707. 10.1016/j.cbi.2019.06.020. [DOI] [PubMed] [Google Scholar]
- Airoldi C.; Sironi E.; Dias C.; Marcelo F.; Martins A.; Rauter A. P.; Nicotra F.; Jimenez-Barbero J. (2013) Natural compounds against Alzheimer’s disease: molecular recognition of Aβ1–42 peptide by Salvia sclareoides extract and its major component, rosmarinic acid, as investigated by NMR. Chem. - Asian J. 8 (3), 596–602. 10.1002/asia.201201063. [DOI] [PubMed] [Google Scholar]
- Levites Y.; Amit T.; Mandel S.; Youdim M. B. (2003) Neuroprotection and neurorescue against Abeta toxicity and PKC-dependent release of nonamyloidogenic soluble precursor protein by green tea polyphenol (−)-epigallocatechin-3-gallate. FASEB J. 17 (8), 952–954. 10.1096/fj.02-0881fje. [DOI] [PubMed] [Google Scholar]
- Baptista F. I.; Henriques A. G.; Silva A. M. S.; Wiltfang J.; da Cruz e Silva O. A. B. (2014) Flavonoids as therapeutic compounds targeting key proteins involved in Alzheimer’s disease. ACS Chem. Neurosci. 5 (2), 83–92. 10.1021/cn400213r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimmyo Y.; Kihara T.; Akaike A.; Niidome T.; Sugimoto H. (2008) Flavonols and flavones as BACE-1 inhibitors: structure-activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim. Biophys. Acta, Gen. Subj. 1780 (5), 819–825. 10.1016/j.bbagen.2008.01.017. [DOI] [PubMed] [Google Scholar]
- Gong E. J.; Park H. R.; Kim M. E.; Piao S.; Lee E.; Jo D.-G.; Chung H. Y.; Ha N.-C.; Mattson M. P.; Lee J. (2011) Morin attenuates tau hyperphosphorylation by inhibiting GSK3β. Neurobiol. Dis. 44 (2), 223–230. 10.1016/j.nbd.2011.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spencer J. P. E. (2007) The interactions of flavonoids within neuronal signalling pathways. Genes Nutr. 2 (3), 257–273. 10.1007/s12263-007-0056-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rezai-Zadeh K.; Douglas Shytle R.; Bai Y.; Tian J.; Hou H.; Mori T.; Zeng J.; Obregon D.; Town T.; Tan J. (2009) Flavonoid-mediated presenilin-1 phosphorylation reduces Alzheimer’s disease β-amyloid production. J. Cell. Mol. Med. 13 (3), 574–588. 10.1111/j.1582-4934.2008.00344.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vila-Nova N. S.; Morais S. M.; Falcão M. J. C.; Bevilaqua C. M. L.; Rondon F. C. M.; Wilson M. E.; Vieira I. G. P.; Andrade H. F. (2012) Leishmanicidal and cholinesterase inhibiting activities of phenolic compounds of Dimorphandra gardneriana and Platymiscium floribundum, native plants from Caatinga biome. Pesquisa Veterinária Brasileira 32, 1164–1168. 10.1590/S0100-736X2012001100015. [DOI] [Google Scholar]
- Jung M.; Park M. (2007) Acetylcholinesterase inhibition by flavonoids from Agrimonia pilosa. Molecules 12 (9), 2130–2139. 10.3390/12092130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orhan I.; Kartal M.; Tosun F.; Sener B. (2007) Screening of various phenolic acids and flavonoid derivatives for their anticholinesterase potential. Z. Naturforsch., C: J. Biosci. 62 (11–12), 829–832. 10.1515/znc-2007-11-1210. [DOI] [PubMed] [Google Scholar]
- Remya C.; Dileep K. V.; Tintu I.; Variyar E. J.; Sadasivan C. (2012) Design of potent inhibitors of acetylcholinesterase using morin as the starting compound. Front. Life Sci. 6 (3–4), 107–117. 10.1080/21553769.2013.815137. [DOI] [Google Scholar]
- Katalinić M.; Rusak G.; Domaćinović Barović J.; Sinko G.; Jelić D.; Antolović R.; Kovarik Z. (2010) Structural aspects of flavonoids as inhibitors of human butyrylcholinesterase. Eur. J. Med. Chem. 45 (1), 186–192. 10.1016/j.ejmech.2009.09.041. [DOI] [PubMed] [Google Scholar]
- Xu S. L.; Bi C. W.; Choi R. C.; Zhu K. Y.; Miernisha A.; Dong T. T.; Tsim K. W. (2013) Flavonoids induce the synthesis and secretion of neurotrophic factors in cultured rat astrocytes: a signaling response mediated by estrogen receptor. Evidence-Based Complementary and Alternative Medicine 2013, 1. 10.1155/2013/127075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z.; Liu X.; Schroeder J. P.; Chan C.-B.; Song M.; Yu S. P.; Weinshenker D.; Ye K. (2014) 7,8-dihydroxyflavone prevents synaptic loss and memory deficits in a mouse model of Alzheimer’s disease. Neuropsychopharmacology 39 (3), 638–650. 10.1038/npp.2013.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaba B.; Khan T.; Haider M. F.; Alam T.; Baboota S.; Parvez S.; Ali J. (2019) Vitamin E Loaded Naringenin Nanoemulsion via Intranasal Delivery for the Management of Oxidative Stress in a 6-OHDA Parkinson’s Disease Model. BioMed Res. Int. 2019, 2382563. 10.1155/2019/2382563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Md S.; Gan S. Y.; Haw Y. H.; Ho C. L.; Wong S.; Choudhury H. (2018) In vitro neuroprotective effects of naringenin nanoemulsion against β-amyloid toxicity through the regulation of amyloidogenesis and tau phosphorylation. Int. J. Biol. Macromol. 118, 1211–1219. 10.1016/j.ijbiomac.2018.06.190. [DOI] [PubMed] [Google Scholar]
- Manach C.; Morand C.; Crespy V.; Demigné C.; Texier O.; Régérat F.; Rémésy C. (1998) Quercetin is recovered in human plasma as conjugated derivatives which retain antioxidant properties. FEBS Lett. 426 (3), 331–336. 10.1016/S0014-5793(98)00367-6. [DOI] [PubMed] [Google Scholar]
- Kuo Y.-C.; Tsao C.-W. (2017) Neuroprotection against apoptosis of SK-N-MC cells using RMP-7- and lactoferrin-grafted liposomes carrying quercetin. Int. J. Nanomed. 12, 2857–2869. 10.2147/IJN.S132472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emerich D. F.; Dean R. L.; Marsh J.; Pink M.; Lafreniere D.; Snodgrass P.; Bartus R. T. (2000) Intravenous cereport (RMP-7) enhances delivery of hydrophilic chemotherapeutics and increases survival in rats with metastatic tumors in the brain. Pharm. Res. 17 (10), 1212–1219. 10.1023/A:1026462629438. [DOI] [PubMed] [Google Scholar]
- Rabelo C. A. S.; Taarji N.; Khalid N.; Kobayashi I.; Nakajima M.; Neves M. A. (2018) Formulation and characterization of water-in-oil nanoemulsions loaded with açaí berry anthocyanins: Insights of degradation kinetics and stability evaluation of anthocyanins and nanoemulsions. Food Res. Int. 106, 542–548. 10.1016/j.foodres.2018.01.017. [DOI] [PubMed] [Google Scholar]
- Ali T.; Kim M. J.; Rehman S. U.; Ahmad A.; Kim M. O. (2017) Anthocyanin-Loaded PEG-Gold Nanoparticles Enhanced the Neuroprotection of Anthocyanins in an Aβ(1–42) Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 54 (8), 6490–6506. 10.1007/s12035-016-0136-4. [DOI] [PubMed] [Google Scholar]
- Aliakbari F.; Shabani A. A.; Bardania H.; Mohammad-Beigi H.; Tayaranian Marvian A.; Dehghani Esmatabad F.; Vafaei A. A.; Shojaosadati S. A.; Saboury A. A.; Christiansen G.; Otzen D. E.; Morshedi D. (2018) Formulation and anti-neurotoxic activity of baicalein-incorporating neutral nanoliposome. Colloids Surf., B 161, 578–587. 10.1016/j.colsurfb.2017.11.023. [DOI] [PubMed] [Google Scholar]
- Zhang J.; Zhou X.; Yu Q.; Yang L.; Sun D.; Zhou Y.; Liu J. (2014) Epigallocatechin-3-gallate (EGCG)-stabilized selenium nanoparticles coated with Tet-1 peptide to reduce amyloid-β aggregation and cytotoxicity. ACS Appl. Mater. Interfaces 6 (11), 8475–8487. 10.1021/am501341u. [DOI] [PubMed] [Google Scholar]
- Loureiro J. A.; Andrade S.; Duarte A.; Neves A. R.; Queiroz J. F.; Nunes C.; Sevin E.; Fenart L.; Gosselet F.; Coelho M. A.; Pereira M. C. (2017) Resveratrol and Grape Extract-loaded Solid Lipid Nanoparticles for the Treatment of Alzheimer’s Disease. Molecules 22 (2), 277. 10.3390/molecules22020277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sadegh Malvajerd S.; Azadi A.; Izadi Z.; Kurd M.; Dara T.; Dibaei M.; Sharif Zadeh M.; Akbari Javar H.; Hamidi M. (2019) Brain Delivery of Curcumin Using Solid Lipid Nanoparticles and Nanostructured Lipid Carriers: Preparation, Optimization, and Pharmacokinetic Evaluation. ACS Chem. Neurosci. 10 (1), 728–739. 10.1021/acschemneuro.8b00510. [DOI] [PubMed] [Google Scholar]
- Sasaki H.; Sunagawa Y.; Takahashi K.; Imaizumi A.; Fukuda H.; Hashimoto T.; Wada H.; Katanasaka Y.; Kakeya H.; Fujita M.; Hasegawa K.; Morimoto T. (2011) Innovative preparation of curcumin for improved oral bioavailability. Biol. Pharm. Bull. 34 (5), 660–665. 10.1248/bpb.34.660. [DOI] [PubMed] [Google Scholar]
- Gota V. S.; Maru G. B.; Soni T. G.; Gandhi T. R.; Kochar N.; Agarwal M. G. (2010) Safety and pharmacokinetics of a solid lipid curcumin particle formulation in osteosarcoma patients and healthy volunteers. J. Agric. Food Chem. 58 (4), 2095–2099. 10.1021/jf9024807. [DOI] [PubMed] [Google Scholar]
- Maiti P.; Dunbar G. L. (2017) Comparative Neuroprotective Effects of Dietary Curcumin and Solid Lipid Curcumin Particles in Cultured Mouse Neuroblastoma Cells after Exposure to Aβ42. Int. J. Alzheimer's Dis. 2017, 4164872. 10.1155/2017/4164872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maiti P.; Hall T. C.; Paladugu L.; Kolli N.; Learman C.; Rossignol J.; Dunbar G. L. (2016) A comparative study of dietary curcumin, nanocurcumin, and other classical amyloid-binding dyes for labeling and imaging of amyloid plaques in brain tissue of 5×-familial Alzheimer’s disease mice. Histochem. Cell Biol. 146 (5), 609–625. 10.1007/s00418-016-1464-1. [DOI] [PubMed] [Google Scholar]
- Kakkar V.; Mishra A. K.; Chuttani K.; Kaur I. P. (2013) Proof of concept studies to confirm the delivery of curcumin loaded solid lipid nanoparticles (C-SLNs) to brain. Int. J. Pharm. 448 (2), 354–359. 10.1016/j.ijpharm.2013.03.046. [DOI] [PubMed] [Google Scholar]
- Marslin G.; Siram K.; Maqbool Q.; Selvakesavan R. K.; Kruszka D.; Kachlicki P.; Franklin G. (2018) Secondary Metabolites in the Green Synthesis of Metallic Nanoparticles. Materials 11 (6), 940. 10.3390/ma11060940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng X.; Chen A.; Zhang Y.; Wang J.; Shao L.; Wei L. (2015) Central nervous system toxicity of metallic nanoparticles. Int. J. Nanomed. 10, 4321–4340. 10.2147/IJN.S78308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fukami G.; Hashimoto K.; Koike K.; Okamura N.; Shimizu E.; Iyo M. (2004) Effect of antioxidant N-acetyl-L-cysteine on behavioral changes and neurotoxicity in rats after administration of methamphetamine. Brain Res. 1016 (1), 90–95. 10.1016/j.brainres.2004.04.072. [DOI] [PubMed] [Google Scholar]
- Khurana A.; Tekula S.; Saifi M. A.; Venkatesh P.; Godugu C. (2019) Therapeutic applications of selenium nanoparticles. Biomed. Pharmacother. 111, 802–812. 10.1016/j.biopha.2018.12.146. [DOI] [PubMed] [Google Scholar]
- Yang L.; Sun J.; Xie W.; Liu Y.; Liu J. (2017) Dual-functional selenium nanoparticles bind to and inhibit amyloid β fiber formation in Alzheimer’s disease. J. Mater. Chem. B 5 (30), 5954–5967. 10.1039/C6TB02952C. [DOI] [PubMed] [Google Scholar]
- Yin T.; Yang L.; Liu Y.; Zhou X.; Sun J.; Liu J. (2015) Sialic acid (SA)-modified selenium nanoparticles coated with a high blood-brain barrier permeability peptide-B6 peptide for potential use in Alzheimer’s disease. Acta Biomater. 25, 172–183. 10.1016/j.actbio.2015.06.035. [DOI] [PubMed] [Google Scholar]
- Kuppusamy P.; Yusoff M. M.; Maniam G. P.; Govindan N. (2016) Biosynthesis of metallic nanoparticles using plant derivatives and their new avenues in pharmacological applications - An updated report. Saudi Pharm. J. 24 (4), 473–484. 10.1016/j.jsps.2014.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ovais M.; Zia N.; Ahmad I.; Khalil A. T.; Raza A.; Ayaz M.; Sadiq A.; Ullah F.; Shinwari Z. K. (2018) Phyto-Therapeutic and Nanomedicinal Approaches to Cure Alzheimer’s Disease: Present Status and Future Opportunities. Front. Aging Neurosci. 10, 284. 10.3389/fnagi.2018.00284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonawane S. K.; Ahmad A.; Chinnathambi S. (2019) Protein-Capped Metal Nanoparticles Inhibit Tau Aggregation in Alzheimer’s Disease. ACS Omega 4 (7), 12833–12840. 10.1021/acsomega.9b01411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Migliore L.; Uboldi C.; Di Bucchianico S.; Coppedè F. (2015) Nanomaterials and neurodegeneration. Environmental and Molecular Mutagenesis 56 (2), 149–170. 10.1002/em.21931. [DOI] [PubMed] [Google Scholar]
- Singh R.; Shitiz K.; Singh S.; Jha S.; Singh A. (2018) Evaluation of wound dressing properties of chitin membranes containing nanosilver. Biomedical Physics & Engineering Express 4 (2), 025030. 10.1088/2057-1976/aaa9ca. [DOI] [Google Scholar]
- Singh R.; Lillard J. W. Jr. (2009) Nanoparticle-based targeted drug delivery. Exp. Mol. Pathol. 86 (3), 215–223. 10.1016/j.yexmp.2008.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morales-Zavala F.; Arriagada H.; Hassan N.; Velasco C.; Riveros A.; Álvarez A. R.; Minniti A. N.; Rojas-Silva X.; Muñoz L. L.; Vasquez R.; Rodriguez K.; Sanchez-Navarro M.; Giralt E.; Araya E.; Aldunate R.; Kogan M. J. (2017) Peptide multifunctionalized gold nanorods decrease toxicity of β-amyloid peptide in a Caenorhabditis elegans model of Alzheimer’s disease. Nanomedicine 13 (7), 2341–2350. 10.1016/j.nano.2017.06.013. [DOI] [PubMed] [Google Scholar]
- Amanzadeh E.; Esmaeili A.; Abadi R. E. N.; Kazemipour N.; Pahlevanneshan Z.; Beheshti S. (2019) Quercetin conjugated with superparamagnetic iron oxide nanoparticles improves learning and memory better than free quercetin via interacting with proteins involved in LTP. Sci. Rep. 9 (1), 6876. 10.1038/s41598-019-43345-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma N.; Jha S. (2020) Amorphous nanosilica induced toxicity, inflammation and innate immune responses: A critical review. Toxicology 441, 152519. 10.1016/j.tox.2020.152519. [DOI] [PubMed] [Google Scholar]
- Yang X.; He C.; Li J.; Chen H.; Ma Q.; Sui X.; Tian S.; Ying M.; Zhang Q.; Luo Y.; Zhuang Z.; Liu J. (2014) Uptake of silica nanoparticles: neurotoxicity and Alzheimer-like pathology in human SK-N-SH and mouse neuro2a neuroblastoma cells. Toxicol. Lett. 229 (1), 240–249. 10.1016/j.toxlet.2014.05.009. [DOI] [PubMed] [Google Scholar]
- Bharali D. J.; Klejbor I.; Stachowiak E. K.; Dutta P.; Roy I.; Kaur N.; Bergey E. J.; Prasad P. N.; Stachowiak M. K. (2005) Organically modified silica nanoparticles: a nonviral vector for in vivo gene delivery and expression in the brain. Proc. Natl. Acad. Sci. U. S. A. 102 (32), 11539–11544. 10.1073/pnas.0504926102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nday C. M.; Eleftheriadou D.; Jackson G. (2019) Naringin nanoparticles against neurodegenerative processes: A preliminary work. Hell J. Nucl. Med. 22 (Suppl), 32–41. [PubMed] [Google Scholar]
- Nday C. M.; Halevas E.; Jackson G. E.; Salifoglou A. (2015) Quercetin encapsulation in modified silica nanoparticles: potential use against Cu(II)-induced oxidative stress in neurodegeneration. J. Inorg. Biochem. 145, 51–64. 10.1016/j.jinorgbio.2015.01.001. [DOI] [PubMed] [Google Scholar]
- Halevas E.; Nday C. M.; Salifoglou A. (2016) Hybrid catechin silica nanoparticle influence on Cu(II) toxicity and morphological lesions in primary neuronal cells. J. Inorg. Biochem. 163, 240–249. 10.1016/j.jinorgbio.2016.04.017. [DOI] [PubMed] [Google Scholar]
- Agrawal M.; Ajazuddin; Tripathi D. K.; Saraf S.; Saraf S.; Antimisiaris S. G.; Mourtas S.; Hammarlund-Udenaes M.; Alexander A. (2017) Recent advancements in liposomes targeting strategies to cross blood-brain barrier (BBB) for the treatment of Alzheimer’s disease. J. Controlled Release 260, 61–77. 10.1016/j.jconrel.2017.05.019. [DOI] [PubMed] [Google Scholar]
- Ross C.; Taylor M.; Fullwood N.; Allsop D. (2018) Liposome delivery systems for the treatment of Alzheimer’s disease. Int. J. Nanomed. 13, 8507. 10.2147/IJN.S183117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salvati E.; Re F.; Sesana S.; Cambianica I.; Sancini G.; Masserini M.; Gregori M. (2013) Liposomes functionalized to overcome the blood-brain barrier and to target amyloid-β peptide: the chemical design affects the permeability across an in vitro model. Int. J. Nanomed. 8, 1749–1758. 10.2147/IJN.S42783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Immordino M. L.; Dosio F.; Cattel L. (2006) Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int. J. Nanomed. 1 (3), 297–315. [PMC free article] [PubMed] [Google Scholar]
- Mourtas S.; Canovi M.; Zona C.; Aurilia D.; Niarakis A.; La Ferla B.; Salmona M.; Nicotra F.; Gobbi M.; Antimisiaris S. G. (2011) Curcumin-decorated nanoliposomes with very high affinity for amyloid-β1–42 peptide. Biomaterials 32 (6), 1635–1645. 10.1016/j.biomaterials.2010.10.027. [DOI] [PubMed] [Google Scholar]
- Lazar A. N.; Mourtas S.; Youssef I.; Parizot C.; Dauphin A.; Delatour B.; Antimisiaris S. G.; Duyckaerts C. (2013) Curcumin-conjugated nanoliposomes with high affinity for Aβ deposits: possible applications to Alzheimer disease. Nanomedicine 9 (5), 712–721. 10.1016/j.nano.2012.11.004. [DOI] [PubMed] [Google Scholar]
- Mahmud M.; Piwoni A.; Filiczak N.; Janicka M.; Gubernator J. (2016) Long-Circulating Curcumin-Loaded Liposome Formulations with High Incorporation Efficiency, Stability and Anticancer Activity towards Pancreatic Adenocarcinoma Cell Lines In Vitro. PLoS One 11 (12), e0167787 10.1371/journal.pone.0167787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Priprem A.; Watanatorn J.; Sutthiparinyanont S.; Phachonpai W.; Muchimapura S. (2008) Anxiety and cognitive effects of quercetin liposomes in rats. Nanomedicine 4 (1), 70–78. 10.1016/j.nano.2007.12.001. [DOI] [PubMed] [Google Scholar]
- Swider E.; Koshkina O.; Tel J.; Cruz L. J.; de Vries I. J. M.; Srinivas M. (2018) Customizing poly(lactic-co-glycolic acid) particles for biomedical applications. Acta Biomater. 73, 38–51. 10.1016/j.actbio.2018.04.006. [DOI] [PubMed] [Google Scholar]
- Amin F. U.; Shah S. A.; Badshah H.; Khan M.; Kim M. O. (2017) Anthocyanins encapsulated by PLGA@PEG nanoparticles potentially improved its free radical scavenging capabilities via p38/JNK pathway against Aβ(1–42)-induced oxidative stress. J. Nanobiotechnol. 15 (1), 12. 10.1186/s12951-016-0227-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rakotoarisoa M.; Angelova A. (2018) Amphiphilic Nanocarrier Systems for Curcumin Delivery in Neurodegenerative Disorders. Medicines (Basel) 5 (4), 126. 10.3390/medicines5040126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ezzati Nazhad Dolatabadi J.; Valizadeh H.; Hamishehkar H. (2015) Solid Lipid Nanoparticles as Efficient Drug and Gene Delivery Systems: Recent Breakthroughs. Adv. Pharm. Bull. 5 (2), 151–159. 10.15171/apb.2015.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rostami E.; Kashanian S.; Azandaryani A. H.; Faramarzi H.; Dolatabadi J. E.; Omidfar K. (2014) Drug targeting using solid lipid nanoparticles. Chem. Phys. Lipids 181, 56–61. 10.1016/j.chemphyslip.2014.03.006. [DOI] [PubMed] [Google Scholar]
- Doktorovova S.; Souto E. B.; Silva A. M. (2014) Nanotoxicology applied to solid lipid nanoparticles and nanostructured lipid carriers - a systematic review of in vitro data. Eur. J. Pharm. Biopharm. 87 (1), 1–18. 10.1016/j.ejpb.2014.02.005. [DOI] [PubMed] [Google Scholar]
- Vedagiri A.; Thangarajan S. (2016) Mitigating effect of chrysin loaded solid lipid nanoparticles against Amyloid β25–35 induced oxidative stress in rat hippocampal region: An efficient formulation approach for Alzheimer’s disease. Neuropeptides 58, 111–125. 10.1016/j.npep.2016.03.002. [DOI] [PubMed] [Google Scholar]
- Dhawan S.; Kapil R.; Singh B. (2011) Formulation development and systematic optimization of solid lipid nanoparticles of quercetin for improved brain delivery. J. Pharm. Pharmacol. 63 (3), 342–351. 10.1111/j.2042-7158.2010.01225.x. [DOI] [PubMed] [Google Scholar]
- Rishitha N.; Muthuraman A. (2018) Therapeutic evaluation of solid lipid nanoparticle of quercetin in pentylenetetrazole induced cognitive impairment of zebrafish. Life Sci. 199, 80–87. 10.1016/j.lfs.2018.03.010. [DOI] [PubMed] [Google Scholar]
- Lohcharoenkal W.; Wang L.; Chen Y. C.; Rojanasakul Y. (2014) Protein Nanoparticles as Drug Delivery Carriers for Cancer Therapy. BioMed Res. Int. 2014, 180549. 10.1155/2014/180549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jain A.; Singh S. K.; Arya S. K.; Kundu S. C.; Kapoor S. (2018) Protein Nanoparticles: Promising Platforms for Drug Delivery Applications. ACS Biomater. Sci. Eng. 4 (12), 3939–3961. 10.1021/acsbiomaterials.8b01098. [DOI] [PubMed] [Google Scholar]
- Verma D.; Gulati N.; Kaul S.; Mukherjee S.; Nagaich U. (2018) Protein Based Nanostructures for Drug Delivery. J. Pharm. (N. Y., NY, U. S.) 2018, 9285853–9285854. 10.1155/2018/9285854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pascoli M.; de Lima R.; Fraceto L. F. (2018) Zein Nanoparticles and Strategies to Improve Colloidal Stability: A Mini-Review. Front. Chem. 6, 5–6. 10.3389/fchem.2018.00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moreno L.; Puerta E.; Suárez-Santiago J. E.; Santos-Magalhães N. S.; Ramirez M. J.; Irache J. M. (2017) Effect of the oral administration of nanoencapsulated quercetin on a mouse model of Alzheimer’s disease. Int. J. Pharm. 517 (1–2), 50–57. 10.1016/j.ijpharm.2016.11.061. [DOI] [PubMed] [Google Scholar]
- Penalva R.; Esparza I.; Larraneta E.; González-Navarro C. J.; Gamazo C.; Irache J. M. (2015) Zein-Based Nanoparticles Improve the Oral Bioavailability of Resveratrol and Its Anti-inflammatory Effects in a Mouse Model of Endotoxic Shock. J. Agric. Food Chem. 63 (23), 5603–5611. 10.1021/jf505694e. [DOI] [PubMed] [Google Scholar]
- Peñalva R.; Morales J.; González-Navarro C. J.; Larrañeta E.; Quincoces G.; Peñuelas I.; Irache J. M. (2018) Increased Oral Bioavailability of Resveratrol by Its Encapsulation in Casein Nanoparticles. Int. J. Mol. Sci. 19 (9), 2816. 10.3390/ijms19092816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Upadhyay A.; Joshi V.; Amanullah A.; Mishra R.; Arora N.; Prasad A.; Mishra A. (2017) E3 Ubiquitin Ligases Neurobiological Mechanisms: Development to Degeneration. Front. Mol. Neurosci. 10, 151. 10.3389/fnmol.2017.00151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jellinger K. A. (2010) Basic mechanisms of neurodegeneration: a critical update. J. Cell. Mol. Med. 14 (3), 457–487. 10.1111/j.1582-4934.2010.01010.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Youdim K. A.; Shukitt-Hale B.; Joseph J. A. (2004) Flavonoids and the brain: interactions at the blood-brain barrier and their physiological effects on the central nervous system. Free Radical Biol. Med. 37 (11), 1683–1693. 10.1016/j.freeradbiomed.2004.08.002. [DOI] [PubMed] [Google Scholar]
- Baum L.; Lam C. W.; Cheung S. K.; Kwok T.; Lui V.; Tsoh J.; Lam L.; Leung V.; Hui E.; Ng C.; Woo J.; Chiu H. F.; Goggins W. B.; Zee B. C.; Cheng K. F.; Fong C. Y.; Wong A.; Mok H.; Chow M. S.; Ho P. C.; Ip S. P.; Ho C. S.; Yu X. W.; Lai C. Y.; Chan M. H.; Szeto S.; Chan I. H.; Mok V. (2008) Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J. Clin. Psychopharmacol. 28 (1), 110–113. 10.1097/jcp.0b013e318160862c. [DOI] [PubMed] [Google Scholar]
- Kennedy D. O.; Wightman E. L.; Reay J. L.; Lietz G.; Okello E. J.; Wilde A.; Haskell C. F. (2010) Effects of resveratrol on cerebral blood flow variables and cognitive performance in humans: a double-blind, placebo-controlled, crossover investigation. Am. J. Clin. Nutr. 91 (6), 1590–1597. 10.3945/ajcn.2009.28641. [DOI] [PubMed] [Google Scholar]
- Barua S.; Mitragotri S. (2014) Challenges associated with Penetration of Nanoparticles across Cell and Tissue Barriers: A Review of Current Status and Future Prospects. Nano Today 9 (2), 223–243. 10.1016/j.nantod.2014.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walker J. M.; Klakotskaia D.; Ajit D.; Weisman G. A.; Wood W. G.; Sun G. Y.; Serfozo P.; Simonyi A.; Schachtman T. R. (2015) Beneficial effects of dietary EGCG and voluntary exercise on behavior in an Alzheimer’s disease mouse model. J. Alzheimer's Dis. 44 (2), 561–572. 10.3233/JAD-140981. [DOI] [PubMed] [Google Scholar]
- Guo Y.; Zhao Y.; Nan Y.; Wang X.; Chen Y.; Wang S. (2017) (−)-Epigallocatechin-3-gallate ameliorates memory impairment and rescues the abnormal synaptic protein levels in the frontal cortex and hippocampus in a mouse model of Alzheimer’s disease. NeuroReport 28 (10), 590–597. 10.1097/WNR.0000000000000803. [DOI] [PubMed] [Google Scholar]
- Rezai-Zadeh K.; Arendash G. W.; Hou H.; Fernandez F.; Jensen M.; Runfeldt M.; Shytle R. D.; Tan J. (2008) Green tea epigallocatechin-3-gallate (EGCG) reduces beta-amyloid mediated cognitive impairment and modulates tau pathology in Alzheimer transgenic mice. Brain Res. 1214, 177–187. 10.1016/j.brainres.2008.02.107. [DOI] [PubMed] [Google Scholar]
- Gu H. F.; Nie Y. X.; Tong Q. Z.; Tang Y. L.; Zeng Y.; Jing K. Q.; Zheng X. L.; Liao D. F. (2014) Epigallocatechin-3-gallate attenuates impairment of learning and memory in chronic unpredictable mild stress-treated rats by restoring hippocampal autophagic flux. PLoS One 9 (11), e112683 10.1371/journal.pone.0112683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vepsäläinen S.; Koivisto H.; Pekkarinen E.; Mäkinen P.; Dobson G.; McDougall G. J.; Stewart D.; Haapasalo A.; Karjalainen R. O.; Tanila H.; Hiltunen M. (2013) Anthocyanin-enriched bilberry and blackcurrant extracts modulate amyloid precursor protein processing and alleviate behavioral abnormalities in the APP/PS1 mouse model of Alzheimer’s disease. J. Nutr. Biochem. 24 (1), 360–370. 10.1016/j.jnutbio.2012.07.006. [DOI] [PubMed] [Google Scholar]
- Giri R. K.; Rajagopal V.; Kalra V. K. (2004) Curcumin, the active constituent of turmeric, inhibits amyloid peptide-induced cytochemokine gene expression and CCR5-mediated chemotaxis of THP-1 monocytes by modulating early growth response-1 transcription factor. J. Neurochem. 91 (5), 1199–1210. 10.1111/j.1471-4159.2004.02800.x. [DOI] [PubMed] [Google Scholar]
- Park S.-Y.; Kim D. S. H. L. (2002) Discovery of Natural Products from Curcuma longa that Protect Cells from Beta-Amyloid Insult: A Drug Discovery Effort against Alzheimer’s Disease. J. Nat. Prod. 65 (9), 1227–1231. 10.1021/np010039x. [DOI] [PubMed] [Google Scholar]
- Onozuka H.; Nakajima A.; Matsuzaki K.; Shin R.-W.; Ogino K.; Saigusa D.; Tetsu N.; Yokosuka A.; Sashida Y.; Mimaki Y.; Yamakuni T.; Ohizumi Y. (2008) Nobiletin, a Citrus Flavonoid, Improves Memory Impairment and Aβ Pathology in a Transgenic Mouse Model of Alzheimer’s Disease. J. Pharmacol. Exp. Ther. 326 (3), 739–744. 10.1124/jpet.108.140293. [DOI] [PubMed] [Google Scholar]
- Nakajima A.; Aoyama Y.; Shin E. J.; Nam Y.; Kim H. C.; Nagai T.; Yokosuka A.; Mimaki Y.; Yokoi T.; Ohizumi Y.; Yamada K. (2015) Nobiletin, a citrus flavonoid, improves cognitive impairment and reduces soluble Aβ levels in a triple transgenic mouse model of Alzheimer’s disease (3XTg-AD). Behav. Brain Res. 289, 69–77. 10.1016/j.bbr.2015.04.028. [DOI] [PubMed] [Google Scholar]
- Nakajima A.; Aoyama Y.; Nguyen T. T.; Shin E. J.; Kim H. C.; Yamada S.; Nakai T.; Nagai T.; Yokosuka A.; Mimaki Y.; Ohizumi Y.; Yamada K. (2013) Nobiletin, a citrus flavonoid, ameliorates cognitive impairment, oxidative burden, and hyperphosphorylation of tau in senescence-accelerated mouse. Behav. Brain Res. 250, 351–360. 10.1016/j.bbr.2013.05.025. [DOI] [PubMed] [Google Scholar]
- Matsuzaki K.; Yamakuni T.; Hashimoto M.; Haque A. M.; Shido O.; Mimaki Y.; Sashida Y.; Ohizumi Y. (2006) Nobiletin restoring β-amyloid-impaired CREB phosphorylation rescues memory deterioration in Alzheimer’s disease model rats. Neurosci. Lett. 400 (3), 230–234. 10.1016/j.neulet.2006.02.077. [DOI] [PubMed] [Google Scholar]
- Moussa C.; Hebron M.; Huang X.; Ahn J.; Rissman R. A.; Aisen P. S.; Turner R. S. (2017) Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J. Neuroinflammation 14 (1), 0–1. 10.1186/s12974-016-0779-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang S. Q.; Obregon D.; Ehrhart J.; Deng J.; Tian J.; Hou H.; Giunta B.; Sawmiller D.; Tan J. (2013) Baicalein reduces β-amyloid and promotes nonamyloidogenic amyloid precursor protein processing in an Alzheimer’s disease transgenic mouse model. J. Neurosci. Res. 91 (9), 1239–1246. 10.1002/jnr.23244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun P., Yin J.-B., Liu L.-H., Guo J., Wang S.-H., Qu C.-H., and Wang C.-X. (2019) Protective role of Dihydromyricetin in Alzheimer’s disease rat model associated with activating AMPK/SIRT1 signaling pathway. Biosci. Rep., 39, (1), , 10.1042/BSR20180902 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paula P.-C.; Angelica Maria S.-G.; Luis C.-H.; Gloria Patricia C.-G. (2019) Preventive Effect of Quercetin in a Triple Transgenic Alzheimer’s Disease Mice Model. Molecules 24 (12), 2287. 10.3390/molecules24122287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabogal-Guáqueta A. M.; Muñoz-Manco J. I.; Ramírez-Pineda J. R.; Lamprea-Rodriguez M.; Osorio E.; Cardona-Gómez G. P. (2015) The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 93, 134–145. 10.1016/j.neuropharm.2015.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kouhestani S.; Jafari A.; Babaei P. (2018) Kaempferol attenuates cognitive deficit via regulating oxidative stress and neuroinflammation in an ovariectomized rat model of sporadic dementia. Neural Regener. Res. 13 (10), 1827–1832. 10.4103/1673-5374.238714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song X.; Liu B.; Cui L.; Zhou B.; Liu W.; Xu F.; Hayashi T.; Hattori S.; Ushiki-Kaku Y.; Tashiro S. I.; Ikejima T. (2017) Silibinin ameliorates anxiety/depression-like behaviors in amyloid β-treated rats by upregulating BDNF/TrkB pathway and attenuating autophagy in hippocampus. Physiol. Behav. 179, 487–493. 10.1016/j.physbeh.2017.07.023. [DOI] [PubMed] [Google Scholar]
- Zhao L.; Wang J.-L.; Liu R.; Li X.-X.; Li J.-F.; Zhang L. (2013) Neuroprotective, anti-amyloidogenic and neurotrophic effects of apigenin in an Alzheimer’s disease mouse model. Molecules 18 (8), 9949–9965. 10.3390/molecules18089949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moghbelinejad S.; Nassiri-Asl M.; Farivar T. N.; Abbasi E.; Sheikhi M.; Taghiloo M.; Farsad F.; Samimi A.; Hajiali F. (2014) Rutin activates the MAPK pathway and BDNF gene expression on beta-amyloid induced neurotoxicity in rats. Toxicol. Lett. 224 (1), 108–113. 10.1016/j.toxlet.2013.10.010. [DOI] [PubMed] [Google Scholar]
- Huang D. S.; Yu Y. C.; Wu C. H.; Lin J. Y. (2017) Protective Effects of Wogonin against Alzheimer’s Disease by Inhibition of Amyloidogenic Pathway. Evidence-based complementary and alternative medicine. eCAM 2017, 3545169. 10.1155/2017/3545169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu Y.; Wang J. (2015) Wogonin increases β-amyloid clearance and inhibits tau phosphorylation via inhibition of mammalian target of rapamycin: potential drug to treat Alzheimer’s disease. Neurol Sci. 36 (7), 1181–1188. 10.1007/s10072-015-2070-z. [DOI] [PubMed] [Google Scholar]
- Currais A.; Prior M.; Dargusch R.; Armando A.; Ehren J.; Schubert D.; Quehenberger O.; Maher P. (2014) Modulation of p25 and inflammatory pathways by fisetin maintains cognitive function in Alzheimer’s disease transgenic mice. Aging Cell 13 (2), 379–390. 10.1111/acel.12185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Currais A.; Farrokhi C.; Dargusch R.; Armando A.; Quehenberger O.; Schubert D.; Maher P. (2018) Fisetin Reduces the Impact of Aging on Behavior and Physiology in the Rapidly Aging SAMP8Mouse. J. Gerontol., Ser. A 73 (3), 299–307. 10.1093/gerona/glx104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heo H. J.; Lee C. Y. (2005) Epicatechin and Catechin in Cocoa Inhibit Amyloid β Protein Induced Apoptosis. J. Agric. Food Chem. 53 (5), 1445–1448. 10.1021/jf048989m. [DOI] [PubMed] [Google Scholar]
- Devi L.; Ohno M. (2012) 7,8-dihydroxyflavone, a small-molecule TrkB agonist, reverses memory deficits and BACE1 elevation in a mouse model of Alzheimer’s disease. Neuropsychopharmacology 37 (2), 434–444. 10.1038/npp.2011.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Z.; Liu X.; Schroeder J. P.; Chan C. B.; Song M.; Yu S. P.; Weinshenker D.; Ye K. (2014) 7,8-dihydroxyflavone prevents synaptic loss and memory deficits in a mouse model of Alzheimer’s disease. Neuropsychopharmacology 39 (3), 638–650. 10.1038/npp.2013.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong E. J.; Park H. R.; Kim M. E.; Piao S.; Lee E.; Jo D. G.; Chung H. Y.; Ha N. C.; Mattson M. P.; Lee J. (2011) Morin attenuates tau hyperphosphorylation by inhibiting GSK3β. Neurobiol. Dis. 44 (2), 223–230. 10.1016/j.nbd.2011.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rezai-Zadeh K.; Douglas Shytle R.; Bai Y.; Tian J.; Hou H.; Mori T.; Zeng J.; Obregon D.; Town T.; Tan J. (2009) Flavonoid-mediated presenilin-1 phosphorylation reduces Alzheimer’s disease beta-amyloid production. J. Cell. Mol. Med. 13 (3), 574–588. 10.1111/j.1582-4934.2008.00344.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sawmiller D.; Habib A.; Li S.; Darlington D.; Hou H.; Tian J.; Shytle R. D.; Smith A.; Giunta B.; Mori T.; Tan J. (2016) Diosmin reduces cerebral Aβ levels, tau hyperphosphorylation, neuroinflammation, and cognitive impairment in the 3xTg-AD mice. J. Neuroimmunol. 299, 98–106. 10.1016/j.jneuroim.2016.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang S. M.; Tsai S. Y.; Lin J. A.; Wu C. H.; Yen G. C. (2012) Cytoprotective effects of hesperetin and hesperidin against amyloid β-induced impairment of glucose transport through downregulation of neuronal autophagy. Mol. Nutr. Food Res. 56 (4), 601–609. 10.1002/mnfr.201100682. [DOI] [PubMed] [Google Scholar]
- Li C.; Zug C.; Qu H.; Schluesener H.; Zhang Z. (2015) Hesperidin ameliorates behavioral impairments and neuropathology of transgenic APP/PS1 mice. Behav. Brain Res. 281, 32–42. 10.1016/j.bbr.2014.12.012. [DOI] [PubMed] [Google Scholar]
- Zhao S. S.; Yang W. N.; Jin H.; Ma K. G.; Feng G. F. (2015) Puerarin attenuates learning and memory impairments and inhibits oxidative stress in STZ-induced SAD mice. NeuroToxicology 51, 166–171. 10.1016/j.neuro.2015.10.010. [DOI] [PubMed] [Google Scholar]
- Zhang H.; Liu Y.; Lao M.; Ma Z.; Yi X. (2011) Puerarin protects Alzheimer’s disease neuronal cybrids from oxidant-stress induced apoptosis by inhibiting pro-death signaling pathways. Exp. Gerontol. 46 (1), 30–37. 10.1016/j.exger.2010.09.013. [DOI] [PubMed] [Google Scholar]
- Zhou Y.; Xie N.; Li L.; Zou Y.; Zhang X.; Dong M. (2014) Puerarin alleviates cognitive impairment and oxidative stress in APP/PS1 transgenic mice. Int. J. Neuropsychopharmacol. 17 (4), 635–644. 10.1017/S146114571300148X. [DOI] [PubMed] [Google Scholar]
- Yang W.; Ma J.; Liu Z.; Lu Y.; Hu B.; Yu H. (2014) Effect of naringenin on brain insulin signaling and cognitive functions in ICV-STZ induced dementia model of rats. Neurol Sci. 35 (5), 741–751. 10.1007/s10072-013-1594-3. [DOI] [PubMed] [Google Scholar]
- Sachdeva A. K.; Kuhad A.; Chopra K. (2014) Naringin ameliorates memory deficits in experimental paradigm of Alzheimer’s disease by attenuating mitochondrial dysfunction. Pharmacol., Biochem. Behav. 127, 101–110. 10.1016/j.pbb.2014.11.002. [DOI] [PubMed] [Google Scholar]
- Pierzynowska K.; Podlacha M.; Gaffke L.; Majkutewicz I.; Mantej J.; Węgrzyn A.; Osiadły M.; Myślińska D.; Węgrzyn G. (2019) Autophagy-dependent mechanism of genistein-mediated elimination of behavioral and biochemical defects in the rat model of sporadic Alzheimer’s disease. Neuropharmacology 148, 332–346. 10.1016/j.neuropharm.2019.01.030. [DOI] [PubMed] [Google Scholar]
- Bonechi C.; Martini S.; Ciani L.; Lamponi S.; Rebmann H.; Rossi C.; Ristori S. (2012) Using liposomes as carriers for polyphenolic compounds: the case of trans-resveratrol. PLoS One 7 (8), e41438 10.1371/journal.pone.0041438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Priprem A.; Watanatorn J.; Sutthiparinyanont S.; Phachonpai W.; Muchimapura S. (2008) Anxiety and cognitive effects of quercetin liposomes in rats. Nanomedicine 4 (1), 70–78. 10.1016/j.nano.2007.12.001. [DOI] [PubMed] [Google Scholar]
- Smith A.; Giunta B.; Bickford P. C.; Fountain M.; Tan J.; Shytle R. D. (2010) Nanolipidic particles improve the bioavailability and alpha-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer’s disease. Int. J. Pharm. 389 (1–2), 207–212. 10.1016/j.ijpharm.2010.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halevas E.; Nday C. M.; Salifoglou A. (2016) Hybrid catechin silica nanoparticle influence on Cu(II) toxicity and morphological lesions in primary neuronal cells. J. Inorg. Biochem. 163, 240–249. 10.1016/j.jinorgbio.2016.04.017. [DOI] [PubMed] [Google Scholar]
- Nday C. M.; Eleftheriadou D.; Jackson G. (2019) Magnetic chrysin silica nanomaterials behavior in an amyloidogenic environment. Hell J. Nucl. Med. 22 (Suppl), 42–50. [PubMed] [Google Scholar]
- Nday C. M.; Halevas E.; Jackson G. E.; Salifoglou A. (2015) Quercetin encapsulation in modified silica nanoparticles: potential use against Cu(II)-induced oxidative stress in neurodegeneration. J. Inorg. Biochem. 145, 51–64. 10.1016/j.jinorgbio.2015.01.001. [DOI] [PubMed] [Google Scholar]
- Huo X.; Zhang Y.; Jin X.; Li Y.; Zhang L. (2019) A novel synthesis of selenium nanoparticles encapsulated PLGA nanospheres with curcumin molecules for the inhibition of amyloid β aggregation in Alzheimer’s disease. J. Photochem. Photobiol., B 190, 98–102. 10.1016/j.jphotobiol.2018.11.008. [DOI] [PubMed] [Google Scholar]
- Cano A.; Ettcheto M.; Chang J. H.; Barroso E.; Espina M.; Kühne B. A.; Barenys M.; Auladell C.; Folch J.; Souto E. B.; Camins A.; Turowski P.; García M. L. (2019) Dual-drug loaded nanoparticles of Epigallocatechin-3-gallate (EGCG)/Ascorbic acid enhance therapeutic efficacy of EGCG in a APPswe/PS1dE9 Alzheimer’s disease mice model. J. Controlled Release 301, 62–75. 10.1016/j.jconrel.2019.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh N. A.; Bhardwaj V.; Ravi C.; Ramesh N.; Mandal A. K. A.; Khan Z. A. (2018) EGCG Nanoparticles Attenuate Aluminum Chloride Induced Neurobehavioral Deficits, Beta Amyloid and Tau Pathology in a Rat Model of Alzheimer’s Disease. Front. Aging Neurosci. 10, 244. 10.3389/fnagi.2018.00244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun D.; Li N.; Zhang W.; Zhao Z.; Mou Z.; Huang D.; Liu J.; Wang W. (2016) Design of PLGA-functionalized quercetin nanoparticles for potential use in Alzheimer’s disease. Colloids Surf., B 148, 116–129. 10.1016/j.colsurfb.2016.08.052. [DOI] [PubMed] [Google Scholar]
- Doggui S.; Sahni J. K.; Arseneault M.; Dao L.; Ramassamy C. (2012) Neuronal uptake and neuroprotective effect of curcumin-loaded PLGA nanoparticles on the human SK-N-SH cell line. J. Alzheimer's Dis. 30 (2), 377–392. 10.3233/JAD-2012-112141. [DOI] [PubMed] [Google Scholar]
- Frozza R. L.; Bernardi A.; Hoppe J. B.; Meneghetti A. B.; Battastini A. M.; Pohlmann A. R.; Guterres S. S.; Salbego C. (2013) Lipid-core nanocapsules improve the effects of resveratrol against Abeta-induced neuroinflammation. J. Biomed. Nanotechnol. 9 (12), 2086–2104. 10.1166/jbn.2013.1709. [DOI] [PubMed] [Google Scholar]
- Loureiro J. A.; Andrade S.; Duarte A.; Neves A. R.; Queiroz J. F.; Nunes C.; Sevin E.; Fenart L.; Gosselet F.; Coelho M. A. N.; Pereira M. C. (2017) Resveratrol and Grape Extract-loaded Solid Lipid Nanoparticles for the Treatment of Alzheimer’s Disease. Molecules 22 (2), 277. 10.3390/molecules22020277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vedagiri A.; Thangarajan S. (2016) Mitigating effect of chrysin loaded solid lipid nanoparticles against Amyloid β25–35 induced oxidative stress in rat hippocampal region: An efficient formulation approach for Alzheimer’s disease. Neuropeptides 58, 111–125. 10.1016/j.npep.2016.03.002. [DOI] [PubMed] [Google Scholar]
- Rishitha N.; Muthuraman A. (2018) Therapeutic evaluation of solid lipid nanoparticle of quercetin in pentylenetetrazole induced cognitive impairment of zebrafish. Life Sci. 199, 80–87. 10.1016/j.lfs.2018.03.010. [DOI] [PubMed] [Google Scholar]
- Tsai M.-J.; Wu P.-C.; Huang Y.-B.; Chang J.-S.; Lin C.-L.; Tsai Y.-H.; Fang J.-Y. (2012) Baicalein loaded in tocol nanostructured lipid carriers (tocol NLCs) for enhanced stability and brain targeting. Int. J. Pharm. 423 (2), 461–470. 10.1016/j.ijpharm.2011.12.009. [DOI] [PubMed] [Google Scholar]
- Kumar P.; Sharma G.; Kumar R.; Singh B.; Malik R.; Katare O. P.; Raza K. (2016) Promises of a biocompatible nanocarrier in improved brain delivery of quercetin: Biochemical, pharmacokinetic and biodistribution evidences. Int. J. Pharm. 515 (1), 307–314. 10.1016/j.ijpharm.2016.10.024. [DOI] [PubMed] [Google Scholar]
- Cheng K. K.; Chan P. S.; Fan S.; Kwan S. M.; Yeung K. L.; Wáng Y. X.; Chow A. H.; Wu E. X.; Baum L. (2015) Curcumin-conjugated magnetic nanoparticles for detecting amyloid plaques in Alzheimer’s disease mice using magnetic resonance imaging (MRI). Biomaterials 44, 155–172. 10.1016/j.biomaterials.2014.12.005. [DOI] [PubMed] [Google Scholar]
- Palmal S.; Maity A. R.; Singh B. K.; Basu S.; Jana N. R.; Jana N. R. (2014) Inhibition of amyloid fibril growth and dissolution of amyloid fibrils by curcumin-gold nanoparticles. Chem. - Eur. J. 20 (20), 6184–6191. 10.1002/chem.201400079. [DOI] [PubMed] [Google Scholar]
- Kim M. J.; Rehman S. U.; Amin F. U.; Kim M. O. (2017) Enhanced neuroprotection of anthocyanin-loaded PEG-gold nanoparticles against Aβ(1–42)-induced neuroinflammation and neurodegeneration via the NF-(K)B /JNK/GSK3β signaling pathway. Nanomedicine 13 (8), 2533–2544. 10.1016/j.nano.2017.06.022. [DOI] [PubMed] [Google Scholar]
- Liu Y.; Zhou H.; Yin T.; Gong Y.; Yuan G.; Chen L.; Liu J. (2019) Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer’s disease. J. Colloid Interface Sci. 552, 388–400. 10.1016/j.jcis.2019.05.066. [DOI] [PubMed] [Google Scholar]