Next Article in Journal
Fluoroquinolone Analogs, SAR Analysis, and the Antimicrobial Evaluation of 7-Benzimidazol-1-yl-fluoroquinolone in In Vitro, In Silico, and In Vivo Models
Next Article in Special Issue
Cannabidiol-Loaded Lipid-Stabilized Nanoparticles Alleviate Psoriasis Severity in Mice: A New Approach for Improved Topical Drug Delivery
Previous Article in Journal
Sustainable Electropolymerization of Zingerone and Its C2 Symmetric Dimer for Amperometric Biosensor Films
Previous Article in Special Issue
Hybrid Nanoplatforms Comprising Organic Nanocompartments Encapsulating Inorganic Nanoparticles for Enhanced Drug Delivery and Bioimaging Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A New Perspective for the Treatment of Alzheimer’s Disease: Exosome-like Liposomes to Deliver Natural Compounds and RNA Therapies

1
Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
2
Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(16), 6015; https://doi.org/10.3390/molecules28166015
Submission received: 15 June 2023 / Revised: 28 July 2023 / Accepted: 1 August 2023 / Published: 11 August 2023

Abstract

:
With the increment of the aging population in recent years, neurodegenerative diseases exert a major global disease burden, essentially as a result of the lack of treatments that stop the disease progression. Alzheimer’s Disease (AD) is an example of a neurodegenerative disease that affects millions of people globally, with no effective treatment. Natural compounds have emerged as a viable therapy to fill a huge gap in AD management, and in recent years, mostly fueled by the COVID-19 pandemic, RNA-based therapeutics have become a hot topic in the treatment of several diseases. Treatments of AD face significant limitations due to the complex and interconnected pathways that lead to their hallmarks and also due to the necessity to cross the blood–brain barrier. Nanotechnology has contributed to surpassing this bottleneck in the treatment of AD by promoting safe and enhanced drug delivery to the brain. In particular, exosome-like nanoparticles, a hybrid delivery system combining exosomes and liposomes’ advantageous features, are demonstrating great potential in the treatment of central nervous system diseases.

1. Introduction

Neurodegenerative diseases are a diversified group of conditions characterized by progressive degeneration of the function of the central nervous system (CNS) or peripheral nervous system, and the current therapeutic options do not provide a cure, only slowing down the disease progression. Together, neurodegenerative diseases exert a major burden in global healthcare systems, with dementia being a public health challenge in many developed countries, as aging is a strong risk factor [1].
Dementia is one of the highest global health crises of this century, with Alzheimer’s disease (AD) being the most common form of dementia. In the United States, an estimated 6.7 million individuals aged 65 and older are living with AD in 2023, and the number is expected to reach 88 million by 2050 [2]. The estimated 2023 cost of caring for those with this disease is $345 billion. Between 2000 and 2019, the number of deaths from AD increased by 145%, while deaths from the number-one cause of death—heart disease—decreased by 7.3% [2].
AD is a progressive, irreversible neurodegenerative disease that leads to memory impairment, impacts cognition, and can ultimately affects behavior, speech, visuospatial orientation, and the motor system. This disease is characterized by two major pathological hallmarks: Progressive accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs). Aβ damages neurons by interfering with neuron communication at synapses and NFTs block the transport of essential molecules for the normal function of neurons. Consequently, these lead to other complications such as oxidative stress, inflammation, and brain atrophy due to cell loss [2,3].
Natural compounds or extracts are a viable therapy to fill the huge gap in the treatment of this disease since they can target several hallmarks. However, for improved efficacy of these compounds, they can be administered by delivery systems. Moreover, mostly instigated by the recent COVID-19 pandemic, RNA-based therapies have become a topic of great interest to researchers and pharmaceutical companies. RNA therapies promise to change the current conventional drugs that are not capable to target and treat all types of diseases, and several clinical studies are ongoing for a variety of RNA-based therapeutics against various incurable diseases. RNA therapy presents several advantages such as cost effectiveness, manufacturing simplicity, and the ability to target previously inaccessible pathways [4,5]. RNA therapies such as microRNAs (miRNAs), small interfering RNAs (siRNAs), and messenger RNAs (mRNAs) seem to be some of the most promising molecules for the treatment of AD [6]. For the effective delivery of RNA, this molecule has to overcome several obstacles. Its hydrophilic, negatively charged properties make it difficult for the RNA molecule to passively diffuse across the cell’s membrane, and so it has to undergo endocytosis and escape from the endosome to reach the cytoplasm. Furthermore, this molecule is highly susceptible to ribonucleases degradation and must have enhanced accumulation at targeted tissues [4].
In addition to the complex mechanisms that lead to AD, the blood–brain barrier (BBB) is known to be a particular reason for the lack of effective treatments for AD. The BBB is a physiological barrier constituted by blood vessels that vascularize the CNS and possess unique properties that allow precise control of the molecules allowed to enter the CNS [7].
Nanoparticles have been used to mitigate all of these hindrances in the delivery of RNA molecules into the brain. These drug delivery systems effectively protect RNA from degradation, enable the crossing of biological barriers, and allow a targeted accumulation and release [8]. In recent years, nanoparticles such as dendrimers, polymeric nanoparticles and gold nanoparticles, and carbon quantum dots have shown to be capable of crossing the BBB effectively [8]. Along with these nanoparticles, exosomes and liposomes are delivery systems with promising properties that allow them to cross the BBB. Exosomes are nano-sized extracellular vesicles (EVs) released into surrounding body fluids by their parental cells and carry cell-specific cargos of proteins, lipids, and genetic materials. These EVs can be selectively uptaken by neighboring or distant cells far from their release. On the other hand, liposomes are synthetic vesicles comprised of one or several concentric lipid bilayers surrounding an aqueous lumen that can be created from cholesterol and natural phospholipids or synthetic surfactants [9,10].
Exosome-like liposomes are a novel concept of nanoparticles that combine the advantages of both the exosomes and liposomes, creating a unique delivery system with several advantages such as the mimetic constituents of natural exosomes, high biocompatibility, small size, easy production, efficient transport and delivery of therapeutical compounds with low bioavailability (e.g., curcumin and RNA molecules), and the ability to load both hydrophilic and hydrophobic drugs [11].

2. Neurodegenerative Diseases

Neurodegenerative diseases are a heterogeneous group of neurological disorders characterized by cognitive, psychiatric, and motor deficits due to neuron loss [12]. In addition to these common features, there are also no current treatments to stop the advancement of the diseases. Some of the main reasons for the lack of effective treatment in neurodegenerative diseases are the limitations imposed by the BBB and the complex pathways that lead to the late diagnosis of the diseases [13].
Neurodegenerative diseases are characterized by (1) protein aggregation; (2) disruptive proteostasis; (3) neuroinflammation; (4) oxidative stress; (5) synaptic failure; and (6) neuronal death (Figure 1). The presence of protein aggregation is a key hallmark in a large variety of neurodegenerative disorders. These abnormally deposited proteins are found in brain regions that, when damaged, lead to physical vulnerability [1,14]. Several proteins are associated with neurodegenerative disorders:
  • Tau protein—microtubule-associated protein—encoded by the microtubule-associated protein tau (MAPT) gene. Tau is substantially expressed in the cytoplasm of neurons and plays an important role primarily in the stabilization and assembly of axonal microtubules and also in a variety of physiological processes, which include axonal transport, signal transmission between neurons, neurogenesis, myelination, motor function, neuronal excitability, glucose metabolism, iron homeostasis, and DNA protection [15,16].
  • Aβ—derives from the amyloid precursor protein (APP) and aggregates into amyloid plaques with Aβ polypeptides 40 and 42 amino acids long [17].
  • Prion—another protein present in neurodegenerative diseases is the prion protein (PrP) encoded by the PRNP gene. In prion diseases, the prion protein misfolds, propagates, and aggregates rapidly, being responsible for spreading neurodegeneration between cells, and, consequently, brain regions [14].
  • α-synuclein—this is a 140-amino-acid protein highly expressed in the brain, encoded by the α-synuclein (SCNA) gene [12].
Under healthy conditions, there are protein degradation systems to maintain protein homeostasis, with an important role in the clearance of toxic protein aggregates: The autophagy lysosomal pathway and the ubiquitin-proteosome system. However, these pathways lose activity in elderly individuals, contributing to the accumulation of toxic protein aggregates [14]. Glial cells (microglia and astrocyte) substitute peripheral immune cells’ function in the brain. Microglia play a crucial role in defense functions in the brain, and these cells are activated in signs of pathogens or injury. In neurotoxic conditions with several aggregated proteins, microglia activation is induced, interacting with astrocytes and leading to inflammation [14,18]. Neurons are particularly susceptible to oxidative stress due to the high polyunsaturated fatty acid content in the membranes, high oxygen consumption, and low antioxidant defenses in the brain, inducing increased oxidation of proteins, nucleic acids, and lipids [1]. Synaptic failure has been described in various neurodegenerative diseases. Synapses are the functional part of the connection between neurons and the key physiological function of neurons. However, when pathogenic factors affect synapses, it suppresses the brain from learning and leads to memory impairment. Currently, these synaptic changes are the targets of many pharmacological interventions [19]. All these factors result in neuronal cell death and, in some neurodegenerative disorders, can result in brain volume loss [14].
Neurodegenerative diseases include Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), and AD, which will be the focus of this review [1].
The brain of PD patients is affected by the presence of intra-neuronal inclusion bodies—Lewy bodies—and the accumulation of the protein α-synuclein, which spreads from one brain region to another. PD is characterized by movement disorder, motor function impairment, and other nonmotor symptoms, such as gastrointestinal issues and sleep disturbances [1,20].
ALS is a devastating, progressive disease, and the cause of this condition is unknown. This disease is characterized by a deficit in motor neurons in the spinal cord and the motor cortex of the cerebrum. Patients witness progressive muscle weakness and atrophy and respiratory failure due to the weakening of respiratory muscles, with a life expectancy of 15.8 months post-diagnosis [21,22].
HD is an inherited neurodegenerative disorder caused by a mutation in the Huntingtin gene, an abnormal trinucleotide expansion, which is translated into a mutant protein. The protein leads to the disruption of cellular molecular processes, which can involve both loss- and gain-of-function mechanisms. A hallmark of this disease is the degeneration of the striatum (caudate nucleus and putamen), with specific loss of efferent medium spiny neurons and brain shrinkage. This disorder is characterized by movement disturbance, cognitive decline, coordination loss, depression, obsessive–compulsive disorder, and other psychiatric symptoms [23,24,25].

Alzheimer’s Disease

The pathological development of AD is complex and not yet fully understood [1]. At the microscopic level, the progression of this disease is characterized by the accumulation of two proteins: Aβ and Tau proteins, which aggregate into Aβ plaques and NFTs, respectively (Figure 2A) [26].
The presence of amyloid plaques is a consequence of an abnormal accumulation and deposition of Aβ, a product of the amyloid precursor protein (APP) [27]. APP is a transmembrane protein expressed in numerous human tissues, including the CNS, and can be cleaved by different proteases in the non-amyloidogenic or amyloidogenic processing of APP [28,29]. If the APP is cleaved by α-secretase, in the non-amyloidogenic pathway, it prevents the formation of the Aβ and, instead, soluble APPα fragments are formed, which are described to be non-cytotoxic [26]. The amyloidogenic pathway includes the combined action of β- and γ- secretases, which generate Aβ peptides with different C-terminal residues (Aβ40 and Aβ42) [15,27]. The deposition and accumulation of these Aβ polypeptides lead to the formation of amyloid plaques in the brain, resulting in neuroinflammation and synaptic dysfunction [30,31,32]. NFTs are a consequence of the deposition of the abnormally phosphorylated Tau protein, leading to its aggregation [16]. The microtubule-binding protein Tau, a cytoskeletal protein produced by alternative splicing of the MAPT gene, has been identified as a key molecule in AD, but also in a series of neurodegenerative diseases referred to as tauopathies, in contrast to Aβ accumulation, which is a characteristic exclusive to AD [15,33,34]. Hyperphosphorylated Tau loses its functions in the synthesis and stabilization of microtubules. These proteins accumulate in neurites and neuronal cell bodies, where it develops into insoluble aggregates, NFTs. Tau can be secreted into the extracellular space either in its naked form or packaged in exosomes [35,36,37].
In addition the accumulation of these two proteins in the brain, there are other hallmarks in AD, such as synaptic impairment neuroinflammation, oxidative stress, and higher activity of acetylcholinesterase (AChE) (Figure 2B). Synaptic impairment is responsible for the reduction in presynaptic vesicle release and the decrease in glutamatergic receptors [33,38]. Additionally, microglia cells develop more susceptibility to stimulus and produce inflammatory cytokines and chemokines, leading to cytotoxic and pro-inflammatory events. Consequently, this contributes to the deterioration of the BBB and the inability to remove neurotoxic molecules such as Aβ plaques and NFTs from the CNS [39,40]. Damage to the mitochondrial structure, integrity, and biogenesis lead to excessive reactive oxygen species (ROS) production, causing damage to the cellular structure. The equilibrium of distinct neurotransmitter systems, such as acetylcholine (Ach), is crucial for healthy brain function [41,42]. In AD, alterations in the cholinergic system are often present, since there is a loss of cholinergic neurons that leads to an extensive decline of ACh, and consequently to a deficit of cholinergic transmission at the pre-synaptic level. This ACh decrease is due to the higher activity of AChE, the enzyme responsible for its degradation [43]. All of the mentioned microscopic hallmarks alter the brain at a macroscopic level, causing brain shrinkage with cortical thinning and atrophy, leading to decreased brain weight [3,27].
Currently, the therapeutical options for these patients are still very limited [44,45]. The main classes of drugs available to treat AD are acetylcholinesterase inhibitors, which include donepezil, rivastigmine, and galantamine. Although the equilibrium of different neurotransmitters, such as acetylcholine, plays a key role in normal brain function, the cholinergic system is not the only system affected by this pathology. Thus, cholinesterase inhibitors only treat symptoms and do not prevent disease progression. Treatments directed to other hallmarks, such as Aβ accumulation and Tau hyperphosphorylation, failed to provide effects [42,46]. Therefore, an innovative approach that can efficiently treat and control each hallmark of this multifactorial disease is required for effective AD treatment [47].

3. Natural Compounds for AD Treatment

Natural drugs are gaining increased interest from both scientific academia and pharmaceutical industries for the therapy of several diseases. In recent years, due to their multiple beneficial properties, more than 100 natural products have been proposed as a promising approach for AD therapy [48]. Many molecules, including lignans, flavonoids, tannins, polyphenols, triterpenes, sterols, and alkaloids, can act via different pathways since they have anti-inflammatory, anti-amyloidogenic, anticholinesterase, and anti-inflammatory properties and can reduce oxidative stress [49]. It is reasonable to speculate that the progression of AD could be slowed down or even prevented by natural products working on multiple pathological targets [50]. Table 1 presents an array of natural compounds or extracts that were researched for AD in the last fifteen years found on PubMed and Web of Science.

Overcoming Limitations of Natural Compounds with Delivery Systems

In addition to the multiple beneficial properties of natural products, these display several limitations such as low hydrophilicity, rapid metabolism and degradation, low bioavailability, reduced targeting, susceptibility to physiological media, and poor permeability through lipid bilayers. Consequently, in vivo, natural drugs require a high-dose administration beyond a safe range, to result in an effective and safe bioavailability [48,79]. Nanotechnology represents a new method to overcome these challenges. Delivery systems for natural compounds lead to the enhancement of pharmacological activity by improving the stability of drugs in vivo, bioavailability, and controlled release, increasing the accumulation of active ingredients in target sites, promoting the solubility of insoluble drugs, and reducing the required doses to produce therapeutic effects [79]. Nanotechnology offers multiple advantages in the delivery of natural products, since by this method, these drugs can exert their therapeutic effect in the treatment of AD (Figure 3) [80].

4. RNAs as a Promising Tool in the Treatment of AD

The small-molecule- and protein-based therapies that interact with a particular biologic molecule to obtain a pharmacological response to control a disease have been successfully dealing with many diseases in past years [81]. However, this conventional pharmacotherapy has several limitations. Protein-based medications primarily target proteins to inhibit their activity, and only ~1.5% of the human genome encodes proteins. Consequently, the range of disease targets of this drug is limited and unable to meet the required demands [5,82]. Most protein-based drugs are too large to enter their target cells and therefore are only effective when their target molecule is extracellular, demonstrating difficult tissue penetration [82,83].
The study of RNA therapeutics started decades ago, leading to a long scientific journey. However, just recently, this field of research has developed dramatically as a result of the response to the COVID-19 pandemic, which revealed how RNA-based therapeutics could lead to a new era of different and accessible new technology in combatting a wide range of diseases [40,84]. In addition to the inherent instability of RNA, these molecules possess particular features and versatility, with multiple advantages over protein and DNA-based drugs. These characteristics include the inducement of protein coding, binding specificity to target molecules, and inhibition of protein translation. Additionally, RNA has the capacity to recognize a wide range of ligands, targeting almost any genetic component within the cell that is out of reach for the most established drug models [81,84].
RNA can be modified in the base, backbone, and sugar, increasing target affinity and preventing nuclease digestion [85]. These modifications make them completely different from cellular RNAs transcribed from the genome [81]. Thus, unlike DNA-based therapeutics, which must cross the cytoplasmic and nuclear membrane and can integrate the host genome and cause a mutation, RNA therapeutics have no risk of chromosomal integration, exhibiting a safer profile [5,86]. Another important advantage of RNA therapy is its long-lasting effects when using, for example, siRNAs as a drug, benefiting patients who cannot receive frequent treatments [87]. The fast production of RNA-based therapies is also a distinct advantage, considering the increased knowledge that will provide faster and easier design of RNA molecules when compared to the process to produce novel protein-based drugs, which takes years. The vaccine’s rapid production and successful reduction of the severity of the disease in infected people during the COVID-19 pandemic is evidence of how quickly this type of therapy can be developed and implemented [86].
A long scientific journey has led to prominent technological advances in the RNA field, and several new types of RNA molecules have been discovered, leading to the development of several studies designed to implement more effective treatments for diseases that still remain without any cure [40]. RNA-based therapeutics can be classified into five different categories: (1) mrna, which encodes for proteins; (2) siRNA, which are double-stranded and primarily cause translational repression of their target protein; (3) mRNAs, which are small RNAs that can either inhibit protein synthesis when they bind to an mRNA target (miRNA mimics) or free up mRNA by binding to the miRNA that represses the translation of that particular mRNA (miRNA inhibitors); (4) antisense oligonucleotides, which are small (~15–25 nucleotides) single-stranded RNAs that can either promote or repress target expression; and (5) aptamers, which are short single-stranded nucleic acids that form secondary and tertiary structures that inhibit several types of target molecules, including proteins [83,84,88].
The use of nucleic acid therapy has limitless potential to treat not only neurological diseases but also a considerable range of disorders since preclinical studies in cellular and animal models proved that mRNAs and short RNAs can be a new class of medicine [89]. Considering the unmet need for effective treatments for AD patients, it is crucial to evaluate diverse therapeutic targets and strategies to cure this disease [44]. In recent years, Aβ and Tau proteins had been the principal targets for researchers. Despite being well-documented hallmarks of AD, treatments involving the regulation of these proteins, unfortunately, are still an unsuccessful strategy [45,90]. In this review, we are going to focus on RNA therapies such as siRNA, miRNA, and mRNA for the treatment of AD (Figure 4) and describe some examples in Table 2.
  • miRNA
miRNAs are small non-coding single-stranded RNAs approximately ~22 nucleotides in length [91]. Endogenous miRNAs are essential in cell development and play a key role in post-transcriptional gene regulation since they regulate the expression of multiple mRNAs both by promoting mRNA degradation and blocking the translation of multiple target mRNAs, inhibiting protein synthesis [84]. They can regulate mRNA translation by binding to the 3′untranslated region, allowing the reduction in the amount of target protein, instead of only inhibiting its activity [40,92]. In addition to gene expression regulation, miRNA can also act as signaling molecules for intercellular communication, revealing that it can be packaged into exosomes to exert this function [93]. The miRNA therapeutic strategy could be categorized into two types: miRNA mimics and miRNA inhibitors [5,82]. miRNA mimics are synthetic RNA molecules that are designed to act as endogenous miRNA to silence genes and can be applied when increased levels of mRNA are prevalent. In contrast, miRNA inhibitors are synthetic ssRNA molecules and can interrupt the miRNA function via sequence-specific binding to mature miRNA, without causing gene silencing. This option is interesting when protein synthesis restoration is needed [82]. These particular features allow miRNA to target multiple sites of various molecularly deregulated cascades in disease conditions, similar to what occurs in AD [40,91]. miRNAs molecules have crucial functions in the nervous system, such as neuronal differentiation, neurite outgrowth, and synaptic plasticity, and are responsive to neuropathological processes, including oxidative stress, neuroinflammation, and protein aggregation. This proves that miRNAs are key molecules in AD, and the dysfunction of miRNAs in this neurological disease is being recognized [45,91].
  • siRNA
siRNAs are short, synthetic double-stranded RNA oligonucleotides (20–25 nt) that take advantage of the RNA interference pathway to silence gene expression by targeting their complementary mRNA [83,84]. siRNAs offer promising therapeutics for brain disease treatment by directly blocking causative gene expression with high targeting specificity, requiring low effective dosages, and benefiting from a relatively simple drug development process [94]. Several siRNA-based therapeutics were already approved by the FDA for other diseases, supporting their potential use for AD therapeutics [40].
  • mRNA
mRNA is a type of single-stranded RNA involved in protein synthesis. The role of mRNA is to carry protein information from the DNA in a cell’s nucleus to the cell’s cytoplasm. Compared to other RNA therapies, mRNA can provide advantages such as (1) safety, (2) effectiveness, particularly in slowly dividing or non-dividing cells such as neural cells, and (3) better control of protein expression [40].
Table 2. miRNA, siRNA, and mRNA therapeutic applications in Alzheimer’s disease.
Table 2. miRNA, siRNA, and mRNA therapeutic applications in Alzheimer’s disease.
Role in ADReferences
Types of miRNA
miR-101Significantly reduced the expression of a reporter under control of APP 3′-UTR in HeLa cells.[95]
miR-106bOverexpression of miR-106b inhibited Aβ1-42-induced tau phosphorylation at Tyr18 in SH-SY5Y cells stably expressing Tau.[96]
miR-137miR-137 inhibited increased expression levels of p-tau induced by Aβ1-42 in SH-SY5Y and inhibited the hyperphosphorylation of Tau protein in a transgenic mouse model of AD. [97]
miR-219In a Drosophila model that produces human Tau, reduction of miR-219 exacerbated Tau toxicity, while overexpression of miR-219 partially annulled toxic effects.[98]
miR-17miR-17 inhibits elevated miR-17 in adult AD (5xFAD) mice microglia improves Aβ degradation.[99]
miR-20b-5pTreatment with miR-20b-5p reduced APP mRNA and protein levels in cultured human neuronal cells.[100]
miR-29cOver-expression of miR-29c in SH-SY5Y, HEK-293T cell lines and miR-29c in transgenic mice downregulated BACE1 protein levels.[101]
miR-298miR-298 is a repressor of APP, BACE1, and the two primary forms of Aβ (Aβ40 and Aβ42) in a primary human cell culture model. Thus, miR-298 significantly reduced levels of ~55 and 50 kDa forms of the Tau protein without significant alterations of total Tau or other forms.[102]
miR-485-5p miR-485-5p overexpression facilitated the learning and memory capabilities of APP/PS1 mice and promoted pericyte viability and prohibited pericyte apoptosis in this model.[103]
miR-9-5pmiR-9-5p overexpression inhibited Aβ25-35-induced mitochondrial dysfunction, cell apoptosis, and oxidative stress by regulating GSK-3β expression in HT22 cells.[104]
miR-132miR-132 inhibited hippocampal iNOS expression and oxidative stress by inhibiting MAPK1 expression to improve the cognitive function of rats with AD.[105]
miR-153Using miR-153 transgenic mouse model, was verified that miR-153 downregulated the expression of APP and APLP2 protein in vivo.[106]
Targeted gene silencing by siRNA
TausiRNA against MAPT can effectively suppress tau expression in vitro and in vivo without a specific delivery agent.[107]
BACE1Polymeric siRNA nanomedicine targeting BACE1 in APP/PS1 transgenic AD mouse model can efficiently penetrate the BBB via glycemia-controlled glucose transporter-1–mediated transport, ensuring that siRNAs decrease BACE1 expression. [94]
Presenilin1 (PS1)Downregulation of PS1 and Aβ42 in IMR32 cells transfected with siRNA against PS1 was verified.[108]
APPInfusion of siRNAs that down-regulated mouse APP protein levels into the ventricular system for 2 weeks down-regulated APP mRNA in mouse brain.[109]
Proteins encoded by mRNA
mRNA encoding neprilysin Neprilysin plays a major role in the clearance of Aβ in the brain. New mRNA therapeutic strategy utilizing mRNA encoding the mouse neprilysin protein has been shown to decrease Aβ deposition and prevent pathogenic changes in the brain.[110]

Overcoming Limitations of RNA Therapies with Delivery Systems

In spite of being a hot research topic in the present day, the development of novel RNA therapeutics has proven to be highly challenging in the past two decades. Some of the major disadvantages that stop RNA from being a clinical success are its instability for in vivo application since the human body has several intrinsic defense systems to protect the cells against exogenous molecules, such as ribonucleases (RNases). RNA’s low targeted tissue accumulation decreases its therapeutic efficacy, requiring high therapeutic RNA doses that can induce toxicity [111]. Early degradation of naked RNAs is another hurdle for their therapeutic efficiency since they tend to be promptly eliminated from the body via renal or hepatic clearance, shortly after the systemic administration [82,111]. For reference, the half-life of naked siRNA is approximately 15 min, and the half-life of naked mRNA can vary between 2 and 25 min, depending on the presence of 5′ capping, the length of the 3′ poly-A tail, and the RNA secondary structure [112]. Finally, being a large and negatively charged molecule, it is difficult to deliver into the cellular cytoplasm, where it exerts its action. Even if cellular internalization of RNA occurs, there is also the risk that RNA cannot escape the endosomal pathway, with only, for example, ~1–2% of siRNAs uptaken by the cells escaping the endosome [81,82].
These obstacles have been considerably surpassed, thanks to the recent advancements in research areas such as RNA biology and nanotechnology that allowed the development of new materials and technologies for the delivery of RNA molecules [81,84]. These new advances transformed RNA technology into a novel therapeutic too since RNA can now be safely transported and delivered to the target thanks to delivery systems [82] (Figure 5).
RNA delivery is key for the treatment of diseases such as neurological disorders, as it gives RNA the ability to target diseases that cannot be treated with other conventional drug groups by encapsulating these molecules in delivery vectors. These RNA vectors have the capacity to effectively protect this molecule from biodegradation, increase bioavailability, solubility and permeation, surpass biological barriers, and promote targeted delivery and release. Hence, RNA nanoencapsulation potentiates the treatment of diseases by silencing genes or expressing therapeutic proteins [113,114].

5. Nanoparticles and the BBB

Delivery systems can overcome RNA’s limitations, becoming a successful therapy, particularly for facilitating the crossing of the BBB, which is the main culprit for the shortage of new and effective treatments for AD [115].
In this review, we will explain the BBB anatomic composition and characteristics, as well as the BBB pathways into the CNS. Subsequently, we will discuss exosomes, liposomes, and exosome-like liposomes as possible tools for RNA transport across the BBB.
From an anatomical point of view, the BBB is composed of different cell types such as endothelial cells, pericytes, and astrocytes (Figure 6A) [116]. In between the endothelial cells, there are tight junctions, which are surrounded by a thin basal membrane and astrocytes vascular feet. These highly restrictive tight junctions are a key BBB feature since they are responsible for the barrier properties and limit the transfer of almost all drugs [116,117]. Pericytes cover 20% of the outer surface of endothelial cells and are responsible for the regulation of the blood flow in the brain capillary through contraction and relaxation. The astrocytes are glial cells that connect the brain capillary and neurons and also maintain BBB functions by providing nutrients to neurons and protecting the brain from oxidative stress and metal toxicity [117]. Additionally, the basal membrane provides structural support around the pericytes and endothelial cells [118].
This specialized barrier acts as an interface with the capacity to regulate the entry of plasma components, red blood cells, and leukocytes into the CNS and ensures the export of potentially neurotoxic molecules from the brain to the blood [119,120]. This barrier strictly controls the molecule movements between the blood and the brain, regulating the homeostasis of the nervous system [119]. Moreover, more than 98% of all small-molecule drugs and approximately 100% of biological drugs are incapable of crossing the BBB. Additionally, water-soluble molecules in the blood are prevented from entering the CNS, while lipid-soluble molecules are reduced by enzymes or efflux pumps [115,121].
The BBB possesses several permanently active transport mechanisms to ensure the transport of nutrients into the CNS while excluding blood-borne molecules that could be detrimental [122]. On one hand, these BBB properties are proof of its vital role in maintaining the specialized microenvironment of the brain tissue. On the other hand, these features make CNS access one of the most difficult of the body, limiting the development of novel effective drugs to treat AD [119]. Even though the BBB is a strict barrier for the circulation of molecules between the blood and the CNS, there are a few pathways that allow the delivery of essential molecules that maintain brain homeostasis (Figure 5B). These include the transcellular pathway, the paracellular pathway, efflux pumps, carrier-mediated transcytosis, receptor-mediated transcytosis, and adsorptive-mediated transcytosis [114].
In a healthy BBB, transcellular diffusion (Figure 6B1) consists of the diffusion of solute particles through the endothelial cells. Particles transported through this route are small lipophilic molecules that penetrate through the cells. On the other hand, paracellular transport (Figure 5B is restricted by the tight junctions between the endothelial cells, allowing only hydrophilic molecules to pass, with a molecular weight < 500 Da.
Efflux pumps (Figure 6B3) are a set of proteins responsible for limiting the accumulation of various potentially toxic molecules, and eventually, for expelling these molecules from the brain. These proteins are a limiting factor for the delivery of bioactive compounds to the brain.
Carrier-mediated transcytosis (Figure 6B4) consists of active transport with the support of carrier proteins such as the glucose transporter isoform (GLUT-1) and the large amino acid transporter (LAT), allowing entrance to glucose or amino acids. The transport of these molecules occurs when they bind to the protein on the blood side of the BBB, and a subsequent conformational change allows their transport into the brain [114,123].
Receptor-mediated transcytosis (Figure 6B5) is a specialized transport system by which endogenous molecules can cross the BBB through receptors present on the cell surface. This type of transport relies on the following mechanisms: Endocytosis, intracellular vesicular trafficking, and exocytosis. Active components bind to their specific receptors on the luminal side of the endothelial cells, and an intracellular vesicle is formed through membrane invagination. The formed vesicles cross the cell to release the ligand at the basolateral side via exocytosis. The most common receptors involved in this process are the transferrin receptor (TfR), the insulin and insulin-like growth factor receptors, the low-density lipoprotein receptor (LDLR), the low-density lipoprotein-receptor-related protein 1 and 2 (LRP1 and LRP2), the scavenger receptor class B type I (SR-B1), the leptin receptor, the albumin receptor, and the lactoferrin receptor. Receptor-mediated transcytosis is one of the most promising pathways for nanoparticle drug delivery through the BBB [114,122].
Adsorptive-mediated transcytosis (Figure 6B6) is another important BBB-crossing pathway, without the involvement of specific plasma-membrane receptors. The basic mechanism is responsible for the transport of charged particles by taking advantage of the electrostatic interactions between the positively charged drug carriers and the negatively charged luminal membrane of the brain endothelial cells. This transport has lower affinity but higher capacity compared to receptor-mediated transcytosis [114,122,123].
There is a crucial need for an ideal and safe approach to effectively carry pharmaceutical agents in a target-specific and sustained-release manner into the CNS, without disrupting the BBB [122,123]. A promising approach is taking advantage of receptor-mediated transcytosis for drug delivery to the brain with the help of ligand-functionalized nanoparticles. Nanoparticles are gaining popularity as drug carriers for the treatment of neurological disorders, due to their small size and unique physical properties [124]. By virtue of their biochemical composition, lipidic nanoparticles provide biomimicking and bio-degradable platforms. As a result of their lipophilicity and size, nanosized particles such as exosomes and liposomes are promising drug delivery carriers to increase the penetration of the BBB [113,125].

5.1. Exosomes

Exosomes are a subset of EVs with a diameter ranging from 30 to 100 nm, and their composition includes lipids, proteins, and nucleic acids [126,127]. Their lipid content includes sphingomyelin, phosphatidylserine, cholesterol, and ceramide or derivatives. Exosomes also carry non-specific proteins (e.g., cytoplasmic enzymes, cytosolic proteins, heat shock proteins, and transferring proteins) and specific proteins that differ from one exosome to another, depending on their origin. The genetic material of these vesicles includes microRNAs, mRNAs, long non-coding RNAs, and DNA fragments [126]. Exosomes are also enriched in late endosome components such as CD63, CD9, and CD81 since they originate from the endocytic compartment of the producer cell in a process that generates multi-vesicular endosomes (MVEs), which subsequently fuse with the plasma membrane to release exosomes into the extracellular space.
Exosomes yield information that can reflect the phenotype of the parental cell since these vesicles carry distinct RNA and protein cargoes that allow the identification of their parental cells, as well as cell-specific or tissue-specific factors that can be used to determine their site of origin [128,129]. Moreover, when exosomes are secreted, undesirable proteins and other molecules are discarded, making these vesicles a compartment of cellular debris for subsequent disposal [130].
Originally, exosomes were primarily described as being for the elimination of excessive and unnecessary molecules from the cells. However, in the last decade, it has been shown that they have other key functions in both physiological and pathological processes. Regarding the physiological roles, exosomes play an important role in intercellular communication since they are able to deliver a number of bioactive cargos to near or distant target cells [126]. Moreover, exosomes display a role in tissue homeostasis and have anti-inflammatory functions. An example of their function is neuronal communication via the secretion of exosomes, which can contribute to a range of neurobiological functions, including synaptic plasticity. In relation to pathological functions, they control the expansion and progression of diseases, such as cancer and neurodegenerative diseases [130]. Exosomes are involved in the complex mechanisms of secretion, spread, and degradation of the Aβ and Tau proteins and are, especially, involved in Tau propagation between neuronal cells [131].
The unique properties of exosomes include their small size, durability, stability, potential cell selectivity, low immunogenicity, and ideal biocompatibility. These properties make them good candidates to be a therapeutic delivery system. Compared to traditional therapeutic drugs, exosomes have a higher potential to pass through the BBB, which helps the drugs they might carry to reach the CNS. Moreover, since exosomes can be isolated from all body fluids, they can be candidates for analysis as part of a non-invasive liquid biopsy [129,130,131].

5.2. Liposomes

In the last two decades, lipid-based nanoparticles (LNPs), especially liposomes, have been attractive nanometric delivery systems for being the most well-studied nonviral platforms for the delivery of RNA molecules and achieving significant clinical success [113,132] highlighted by the highly effective mRNA COVID-19 vaccines [132,133].
There are several types of LNPs such as liposomes, niosomes, transfersomes, nanoemulsions, solid lipid nanoparticles (SLNs), lipid nanocapsules (LNCs), nanostructured lipid carriers (NLCs), lipid-based micelles, core–shell lipid nanoparticles (CLNs), and hybrid lipid-polymeric nanoparticles [132,134]. LNPs range in size from 40 nm to 1000 nm and are colloidal lipophilic systems constituted by four main components: A pH-sensitive cationic lipid, a helper lipid, cholesterol, and a PEG-lipid. The cationic lipid is a synthetic lipid, constituted by a hydrophilic head with a protonable tertiary amino group (pKa 6–6.7) and a long hydrophobic tail. Cholesterol is incorporated into the LNPs formulation with the goal of increasing their flexibility, whilst the helper phospholipid assists in the process of endosomal escape and contributes to the stability of LNPs [135,136]. Finally, the insertion of a short-chain PEG-lipid derivative (normally of 14 carbon atoms) is essential to maximize the ex vivo stability and control the particle size before administration [137].
Due to their resemblance to biological and natural components, these nano systems show tremendous promise as carriers for therapeutic applications. The main advantages of LNPs over other nanoparticles are their low toxicity and biocompatibility, biodegradability, safety, high mechanical and chemical versatility, and the capacity to protect the active ingredient from degradation processes induced by external factors. Along with these features, these lipid-based nanoparticles can incorporate the delivery of both hydrophobic and hydrophilic molecules and most of their preparation methods can be easily scaled up. Additionally, because of their lipophilicity, LNPs possess the ability to overcome difficult physiological barriers, such as the BBB, even without surface modification [124,134].
Among all the nano-based drug delivery systems, liposomes are the most biocompatible and least toxic since they are composed of phospholipids and cholesterol, the main components of cell membranes. Liposomes are an extremely versatile nanocarrier platform that has the capacity to load multiple drugs, provide protection from degradation, have controlled and targeted drug release, and enhance drug endocytosis into cells [11]. Additionally, these nanoparticles are able to incorporate both hydrophilic and hydrophobic therapeutic agents. The hydrophilic compounds may either be entrapped into the aqueous core of the liposomes or be located at the interface between the lipid bilayer and the external water phase, while the hydrophobic compounds are generally entrapped in the hydrophobic core of their lipid bilayers. The positively charged lipids in the liposome’s constitution allow the electrostatic interaction with negatively charged nucleic acids, such as DNA and RNA, in gene delivery applications [138].
There is evidence that lipid-based nanoplatforms will play a key role in the development of RNA neuro-therapies, with liposomes being one of the main lipidic platforms for RNA delivery to the CNS [113]. In order to enhance drug delivery into the CNS, the liposome surface can be modified by the inclusion of biologically active ligands, such as peptides, polysaccharides, antibodies, or aptamers, which specifically bind to receptors expressed on the surface of the brain endothelial cells, facilitating their binding and transport across the BBB. The addition of polyethylene glycol (PEG) offers superficial protection for the liposomes by avoiding binding with plasma proteins, and hence, preventing their opsonization and subsequent clearance. PEGylation of liposomes prolongs their circulation time in the body and also plays a crucial role in brain drug delivery, allowing liposomes to cross the BBB [138].

5.3. Exosome-like Liposomes as a Novel Strategy

Scientists have explored various nanomaterials for targeted delivery through the BBB with significant efficacy, such as dendrimers, polymeric nanoparticles, gold nanoparticles, carbon quantum dots, and exosome-like liposomes [8,139]. First, in this review, we will summarize the exosome-like liposomes’ competition and then focus only on this delivery system.
  • Dendrimers: Dendrimers are highly branched, characterized by defined molecular weights and specific encapsulation properties. This type of delivery system is composed of symmetrical polymeric macromolecules with a large number of reactive surface groups, with three distinctive architectural components: An interior core, an interior layer consisting of repeating units radially attached to the inner core, and functional end groups on the outside layer. Because of these unique features, dendrimers can cross-impair the BBB and target astrocytes and microglia after systemic administration in animal models [139].
  • Polymeric nanoparticles: Polymeric nanoparticles can be produced from synthetic or natural polymers. However, to be applied in brain drug delivery, these nanoparticles need to be biodegradable and biocompatible. PBCA, PLA, and PLGA nanoparticles are nanoparticles able to cross the BBB. These nanocarriers possess controlled drug release, targeting efficiency, and can avoid phagocytosis by the reticuloendothelial system, thus improving the concentration of drugs in the brain [140].
  • Gold nanoparticles: Nanoparticles (mostly < 10 nm in size) composed of a gold core and with covalently or non-covalently attached surface ligands. Multiple in vivo studies on rodents have shown that low amounts of this delivery system were able to cross the BBB. However, greater amounts of the administered dose were found in the liver and in the blood [8]. Additionally, Sela et al. proved that gold nanoparticles could penetrate the BBB of rats without the use of an external field or surface modification and were found to be distributed uniformly in both the hypothalamus and hippocampus indicating there is no selective binding in these regions of the brain [141].
  • Carbon quantum dots: This delivery system retains a polymeric core structure and various functional groups on the surface, facilitating their conjugation with drug molecules for specific delivery. This is a carrier with several efficient features for BBB crossing such as excellent biocompatibility and low toxicity due to the lack of metal elements, small size, and photoluminescence, which can be utilized to track the penetration of CDs through the BBB [117].
In addition to the fact that these nanoparticles are promising in overcoming the BBB, exosome-like liposomes are also delivery systems with unique properties to achieve this goal. Exosomes and liposomes are promising nanocarriers with unique properties. However, these delivery systems have some limitations. For instance, liposomal targeting efficiency is limited, and they can induce immunogenicity [11], whilst efficient and reliable isolation and purification are needed for the clinical application of exosomes. It is also crucial to identify appropriate strategies to increase loading capacity and specificity to use exosomes as a nanocarrier [10,142]. When comparing the advantages and disadvantages of exosomes and liposomes, it becomes evident that the two systems are complementary, since the advantages of the one system mitigate the disadvantages of the other, and vice-versa (Figure 7). Therefore, the development of a system that incorporates the desirable features of the two carriers into one hybrid delivery system, led to an innovative carrier for drug delivery applications, the exosome-like liposome (Figure 7) [11,143]. These novel exosome-like liposomes are formulated with a lipid composition that mimics that of exosomes, which impart them with some of the desirable characteristics of exosomes, such as enhanced passive targeting, biocompatibility and RES evasion, and the ability to cross biological barriers, whilst allowing much higher encapsulation efficiencies and larger-scale production in good manufacturing practice that the use of exosomes themselves does not allow.
Thus, particle size plays a major role in drug delivery efficacy, including in the therapeutic effect achieved. The endothelial cells’ slit width is 200 to 500 nm. Consequently, for the long-term circulation of these nanoparticles, they need to have a size that does not exceed 200 nm, leading to better brain drug delivery across the BBB. Moreover, the fact that exosome-like liposomes are coated with this suitable hydrophilic polymer leads to advantages such as escaping phagocytosis, avoiding opsonization, and further increasing the blood circulation time, since the hydrophobic nature of the nanoparticle containing the drug is shielded. These unique characteristics drive researchers to believe that this type of carrier can have a key impact in crossing the BBB, helping in the therapy of neurodegenerative diseases such as AD [123].
Additionally, exosome-like liposomes were highly efficient at encapsulating curcumin, with encapsulation efficiencies ranging between 85 and 94%, and were effective at delivering curcumin into neuronal cells to promote a superior neuroprotective effect after oxidative insult compared to free curcumin at the same concentrations. Thus, these delivery systems have shown to be highly biocompatible, without significantly affecting cell viability or causing hemolysis. Finally, the exosome-like liposomes have been shown to be internalized by zebrafish embryos and accumulate in lipid-rich zones, such as the brain and yolk sac, also in stages where the BBB is already formed [144]. In Table 3, we demonstrate other crucial examples of applications of exosome-like liposomes in vivo and in vitro.

6. Conclusions

AD is a neurodegenerative disease that has a tremendous impact on people’s quality of life, all around an aging world. This burden is further impacted by the lack of therapies, due to the complex mechanisms that lead to the disease onset and also due to limitations that the BBB imposes on the entrance and maintenance of therapeutic molecules in the CNS.
However, the emergence of RNA as an especially versatile tool for the treatment of several diseases has opened multiple new possibilities for the development of effective treatments for AD.
The accelerated growth of RNA therapies requires the development of efficient delivery systems to transport a selectively deliver the RNA molecules into their target cells or tissues, due to the low bioavailability of these molecules.
Nanoparticles have provided new and safer avenues for the delivery of natural compounds and RNA. In this field, novel exosome-like liposomes are positioning themselves as a prime solution for the vehiculation of therapeutical compounds to the CNS, due to the combination of the favorable characteristics of exosomes with those of liposomes, which entitle scalable production in good manufacturing practices, which is currently unfeasible for the exosomes themselves.

7. Patents

The authors are co-inventors in a patent application (EXOSOME-MIMETIC LIPOSOME COMPOSITION AND USE, Instituto Nacional da Propriedade Industrial PPP No. 116560 P, 3 July 2020).

Author Contributions

Conceptualization, J.R., I.L. and A.C.G.; investigation, formal analysis, J.R.; writing—original draft preparation, J.R.; writing—review and editing, J.R., I.L. and A.C.G.; supervision, I.L. and A.C.G.; funding acquisition, A.C.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the strategic program UID/BIA/04050/2020 funded by National funds through the Fundação para a Ciência e Tecnologia I.P. Ivo Lopes is a recipient of a scholarship from the Direção Regional da Ciência e Tecnologia, Governo Regional dos Açores (M3.1.a/F/128/2015).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data will be made available upon request.

Acknowledgments

We acknowledge Mario Fernandes for support in revisions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bloomingdale, P.; Karelina, T.; Ramakrishnan, V.; Bakshi, S.; Véronneau-Veilleux, F.; Moye, M.; Sekiguchi, K.; Meno-Tetang, G.; Mohan, A.; Maithreye, R.; et al. Hallmarks of neurodegenerative disease: A systems pharmacology perspective. CPT Pharmacomet. Syst. Pharmacol. 2022, 11, 1399–1429. [Google Scholar] [CrossRef]
  2. Alzheimer’s Association. 2023 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2023, 19, 1598–1695. [Google Scholar] [CrossRef]
  3. De Ture, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhu, Y.; Zhu, L.; Wang, X.; Jin, H. RNA-based therapeutics: An overview and prospectus. Cell Death Dis. 2022, 13, 644. [Google Scholar] [CrossRef]
  5. Damase, T.R.; Sukhovershin, R.; Boada, C.; Taraballi, F.; Pettigrew, R.I.; Cooke, J.P. The Limitless Future of RNA Therapeutics. Front. Bioeng. Biotechnol. 2021, 9, 628137. [Google Scholar] [CrossRef] [PubMed]
  6. Luo, M.; Lee, L.K.C.; Peng, B.; Choi, C.H.J.; Tong, W.Y.; Voelcker, N.H. Delivering the Promise of Gene Therapy with Nanomedicines in Treating Central Nervous System Diseases. Adv. Sci. 2022, 9, 2201740. [Google Scholar] [CrossRef]
  7. Daneman, R.; Prat, A. The Blood–Brain Barrier. Cold Spring Harb. Perspect. Biol. 2015, 7, a020412. [Google Scholar] [CrossRef] [Green Version]
  8. Lombardo, S.M.; Schneider, M.; Türeli, A.E.; Günday Türeli, N. Key for crossing the BBB with nanoparticles: The rational design. Beilstein J. Nanotechnol. 2020, 11, 866–883. [Google Scholar] [CrossRef]
  9. Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef] [Green Version]
  10. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
  11. Antimisiaris, S.; Mourtas, S.; Marazioti, A. Exosomes and Exosome-Inspired Vesicles for Targeted Drug Delivery. Pharmaceutics 2018, 10, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Forrest, S.L.; Kovacs, G.G. Current Concepts of Mixed Pathologies in Neurodegenerative Diseases. Can. J. Neurol. Sci. J. Can. Sci. Neurol. 2022, 50, 329–345. [Google Scholar] [CrossRef]
  13. Masoudi Asil, S.; Ahlawat, J.; Guillama Barroso, G.; Narayan, M. Nanomaterial based drug delivery systems for the treatment of neurodegenerative diseases. Biomater. Sci. 2020, 8, 4109–4128. [Google Scholar] [CrossRef] [PubMed]
  14. Wilson, D.M.; Cookson, M.R.; Van Den Bosch, L.; Zetterberg, H.; Holtzman, D.M.; Dewachter, I. Hallmarks of neurodegenerative diseases. Cell 2023, 186, 693–714. [Google Scholar] [CrossRef]
  15. Roda, A.; Serra-Mir, G.; Montoliu-Gaya, L.; Tiessler, L.; Villegas, S. Amyloid-beta peptide and tau protein crosstalk in Alzheimer’s disease. Neural Regen. Res. 2022, 17, 1666. [Google Scholar] [CrossRef]
  16. Liang, S.Y.; Wang, Z.T.; Tan, L.; Yu, J.T. Tau Toxicity in Neurodegeneration. Mol. Neurobiol. 2022, 59, 3617–3634. [Google Scholar] [CrossRef]
  17. Yu, H.; Wu, J. Amyloid-β: A double agent in Alzheimer’s disease? Biomed. Pharmacother. 2021, 139, 111575. [Google Scholar] [CrossRef] [PubMed]
  18. Xiao, Y.; Wang, S.-K.; Zhang, Y.; Rostami, A.; Kenkare, A.; Casella, G.; Yuan, Z.-Q.; Li, X. Role of extracellular vesicles in neurodegenerative diseases. Prog. Neurobiol. 2021, 201, 102022. [Google Scholar] [CrossRef] [PubMed]
  19. Wei, Z.; Wei, M.; Yang, X.; Xu, Y.; Gao, S.; Ren, K. Synaptic Secretion and Beyond: Targeting Synapse and Neurotransmitters to Treat Neurodegenerative Diseases. Oxidative Med. Cell. Longev. 2022, 2022, 9176923. [Google Scholar] [CrossRef]
  20. Van Den Berge, N.; Ulusoy, A. Animal models of brain-first and body-first Parkinson’s disease. Neurobiol. Dis. 2022, 163, 105599. [Google Scholar] [CrossRef]
  21. Sun, X.; Song, J.; Huang, H.; Chen, H.; Qian, K. Modeling hallmark pathology using motor neurons derived from the family and sporadic amyotrophic lateral sclerosis patient-specific iPS cells. Stem Cell Res. Ther. 2018, 9, 315. [Google Scholar] [CrossRef] [Green Version]
  22. Xiong, L.; McCoy, M.; Komuro, H.; West, X.Z.; Yakubenko, V.; Gao, D.; Dudiki, T.; Milo, A.; Chen, J.; Podrez, E.A.; et al. Inflammation-dependent oxidative stress metabolites as a hallmark of amyotrophic lateral sclerosis. Free. Radic. Biol. Med. 2022, 178, 125–133. [Google Scholar] [CrossRef]
  23. Mehler, M.F.; Petronglo, J.R.; Arteaga-Bracho, E.E.; Gulinello, M.E.; Winchester, M.L.; Pichamoorthy, N.; Young, S.K.; DeJesus, C.D.; Ishtiaq, H.; Gokhan, S.; et al. Loss-of-Huntingtin in Medial and Lateral Ganglionic Lineages Differentially Disrupts Regional Interneuron and Projection Neuron Subtypes and Promotes Huntington’s Disease-Associated Behavioral, Cellular, and Pathological Hallmarks. J. Neurosci. 2019, 39, 1892–1909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jimenez-Sanchez, M.; Licitra, F.; Underwood, B.R.; Rubinsztein, D.C. Huntington’s Disease: Mechanisms of Pathogenesis and Therapeutic Strategies. Cold Spring Harb. Perspect. Med. 2017, 7, a024240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Machiela, E.; Southwell, A.L. Biological Aging and the Cellular Pathogenesis of Huntington’s Disease. J. Huntington’s Dis. 2020, 9, 115–128. [Google Scholar] [CrossRef]
  26. Gallego Villarejo, L.; Bachmann, L.; Marks, D.; Brachthäuser, M.; Geidies, A.; Müller, T. Role of Intracellular Amyloid β as Pathway Modulator, Biomarker, and Therapy Target. Int. J. Mol. Sci. 2022, 23, 4656. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, X.Q.; Mobley, W.C. Alzheimer Disease Pathogenesis: Insights from Molecular and Cellular Biology Studies of Oligomeric Aβ and Tau Species. Front. Neurosci. 2019, 13, 659. [Google Scholar] [CrossRef] [PubMed]
  28. Guo, Y.; Wang, Q.; Chen, S.; Xu, C. Functions of amyloid precursor protein in metabolic diseases. Metabolism 2021, 115, 154454. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, X.; Liu, Y.; Ji, S. Secretases Related to Amyloid Precursor Protein Processing. Membranes 2021, 11, 983. [Google Scholar] [CrossRef] [PubMed]
  30. Zhang, T.; Chen, D.; Lee, T.H. Phosphorylation Signaling in APP Processing in Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 21, 209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Lane, C.A.; Hardy, J.; Schott, J.M. Alzheimer’s disease. Eur. J. Neurol. 2018, 25, 59–70. [Google Scholar] [CrossRef] [PubMed]
  32. Beera, A.M.; Seethamraju, S.M.; Nori, L.P. Alzheimer’s Disease: Perspective on Therapeutic Options and Recent Hallmarks in Clinical Research. Int. J. Pharm. Res. Allied Sci. 2021, 10, 110–120. [Google Scholar] [CrossRef]
  33. Wu, M.; Zhang, M.; Yin, X.; Chen, K.; Hu, Z.; Zhou, Q.; Cao, X.; Chen, Z.; Liu, D. The role of pathological tau in synaptic dysfunction in Alzheimer’s diseases. Transl. Neurodegener. 2021, 10, 45. [Google Scholar] [CrossRef] [PubMed]
  34. Chu, D.; Liu, F. Pathological Changes of Tau Related to Alzheimer’s Disease. ACS Chem. Neurosci. 2019, 10, 931–944. [Google Scholar] [CrossRef]
  35. Zhang, H.; Cao, Y.; Ma, L.; Wei, Y.; Li, H. Possible Mechanisms of Tau Spread and Toxicity in Alzheimer’s Disease. Front. Cell Dev. Biol. 2021, 9, 707268. [Google Scholar] [CrossRef]
  36. Fleeman, R.M.; Proctor, E.A. Astrocytic Propagation of Tau in the Context of Alzheimer’s Disease. Front. Cell Neurosci. 2021, 15, 645233. [Google Scholar] [CrossRef]
  37. Silva, M.V.F.; Loures, C.D.M.G.; Alves, L.C.V.; de Souza, L.C.; Borges, K.B.G.; Carvalho, M.D.G. Alzheimer’s disease: Risk factors and potentially protective measures. J. Biomed. Sci. 2019, 26, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Rawat, P.; Sehar, U.; Bisht, J.; Selman, A.; Culberson, J.; Reddy, P.H. Phosphorylated Tau in Alzheimer’s Disease and Other Tauopathies. Int. J. Mol. Sci. 2022, 23, 12841. [Google Scholar] [CrossRef]
  39. DeVos, S.L.; Corjuc, B.T.; Oakley, D.H.; Nobuhara, C.K.; Bannon, R.N.; Chase, A.; Commins, C.; Gonzalez, J.A.; Dooley, P.M.; Frosch, M.P.; et al. Synaptic Tau Seeding Precedes Tau Pathology in Human Alzheimer’s Disease Brain. Front. Neurosci. 2018, 12, 267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Riscado, M.; Baptista, B.; Sousa, F. New RNA-Based Breakthroughs in Alzheimer’s Disease Diagnosis and Therapeutics. Pharmaceutics 2021, 13, 1397. [Google Scholar]
  41. Plascencia-Villa, G.; Perry, G. Neuropathologic Changes Provide Insights into Key Mechanisms of Alzheimer Disease and Related Dementia. Am. J. Pathol. 2022, 192, 1340–1346. [Google Scholar] [CrossRef] [PubMed]
  42. Marucci, G.; Buccioni, M.; Ben, D.D.; Lambertucci, C.; Volpini, R.; Amenta, F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology 2021, 190, 108352. [Google Scholar] [CrossRef] [PubMed]
  43. Vecchio, I.; Sorrentino, L.; Paoletti, A.; Marra, R.; Arbitrio, M. The State of The Art on Acetylcholinesterase Inhibitors in the Treatment of Alzheimer’s Disease. J. Cent. Nerv. Syst. Dis. 2021, 13, 117957352110291. [Google Scholar] [CrossRef]
  44. Bennett, C.F.; Kordasiewicz, H.B.; Cleveland, D.W. Antisense Drugs Make Sense for Neurological Diseases. Annu. Rev. Pharmacol. Toxicol. 2021, 61, 831–852. [Google Scholar] [CrossRef]
  45. Angelucci, F.; Cechova, K.; Valis, M.; Kuca, K.; Zhang, B.; Hort, J. MicroRNAs in Alzheimer’s Disease: Diagnostic Markers or Therapeutic Agents? Front. Pharmacol. 2019, 10, 665. [Google Scholar] [CrossRef]
  46. Noori, T.; Dehpour, A.R.; Sureda, A.; Sobarzo-Sanchez, E.; Shirooie, S. Role of natural products for the treatment of Alzheimer’s disease. Eur. J. Pharmacol. 2021, 898, 173974. [Google Scholar] [CrossRef] [PubMed]
  47. Lee, C.Y.; Ryu, I.S.; Ryu, J.H.; Cho, H.J. miRNAs as Therapeutic Tools in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 13012. [Google Scholar] [CrossRef] [PubMed]
  48. Ramalho, M.J.; Andrade, S.; Loureiro, J.A.; do Carmo Pereira, M. Nanotechnology to improve the Alzheimer’s disease therapy with natural compounds. Drug Deliv. Transl. Res. 2020, 10, 380–402. [Google Scholar] [CrossRef] [PubMed]
  49. Alhazmi, H.A.; Albratty, M. An update on the novel and approved drugs for Alzheimer disease. Saudi Pharm. J. 2022, 30, 1755–1764. [Google Scholar] [CrossRef] [PubMed]
  50. Chen, X.; Drew, J.; Berney, W.; Lei, W. Neuroprotective Natural Products for Alzheimer’s Disease. Cells 2021, 10, 1309. [Google Scholar] [CrossRef] [PubMed]
  51. Said, M.M.; Rabo, M.M.A. Neuroprotective effects of eugenol against aluminiuminduced toxicity in the rat brain. Arch. Ind. Hyg. Toxicol. 2017, 68, 27–37. [Google Scholar] [CrossRef] [Green Version]
  52. Casares, N.; Alfaro, M.; Cuadrado-Tejedor, M.; Lasarte-Cia, A.; Navarro, F.; Vivas, I.; Espelosin, M.; Cartas-Cejudo, P.; Fernández-Irigoyen, J.; Santamaría, E.; et al. Improvement of cognitive function in wild-type and Alzheimer’s disease mouse models by the immunomodulatory properties of menthol inhalation or by depletion of T regulatory cells. Front. Immunol. 2023, 14, 1130044. [Google Scholar] [CrossRef]
  53. Campos, H.M.; da Costa, M.; Moreira, L.K.d.S.; Neri, H.F.d.S.; da Silva, C.R.B.; Pruccoli, L.; dos Santos, F.C.A.; Costa, E.A.; Tarozzi, A.; Ghedini, P.C. Protective effects of chrysin against the neurotoxicity induced by aluminium: In vitro and in vivo studies. Toxicology 2022, 465, 153033. [Google Scholar] [CrossRef] [PubMed]
  54. Hase, T.; Shishido, S.; Yamamoto, S.; Yamashita, R.; Nukima, H.; Taira, S.; Toyoda, T.; Abe, K.; Hamaguchi, T.; Ono, K.; et al. Rosmarinic acid suppresses Alzheimer’s disease development by reducing amyloid β aggregation by increasing monoamine secretion. Sci. Rep. 2019, 9, 8711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Guan, X.; Xu, J.; Liu, J.; Wu, J.; Chen, L. Ginkgo biloba preparation prevents and treats senile dementia by inhibiting neuro-inflammatory responses. Trop. J. Pharm. Res. 2019, 17, 1961. [Google Scholar] [CrossRef]
  56. Islam, F.; Nafady, M.H.; Islam, R.; Saha, S.; Rashid, S.; Akter, A.; Or-Rashid, H.; Akhtar, M.F.; Perveen, A.; Ashraf, G.M.; et al. Resveratrol and neuroprotection: An insight into prospective therapeutic approaches against Alzheimer’s disease from bench to bedside. Mol. Neurobiol. 2022, 59, 4384–4404. [Google Scholar] [CrossRef] [PubMed]
  57. Villegas, C.; Perez, R.; Petiz, L.L.; Glaser, T.; Ulrich, H.; Paz, C. Ginkgolides and Huperzine A for complementary treatment of Alzheimer’s disease. IUBMB Life 2022, 74, 763–779. [Google Scholar] [CrossRef]
  58. Dubey, T.; Chinnathambi, S. Brahmi (Bacopa monnieri): An ayurvedic herb against the Alzheimer’s disease. Arch. Biochem. Biophys. 2019, 676, 108153. [Google Scholar] [CrossRef]
  59. Snow, A.D.; Castillo, G.M.; Nguyen, B.P.; Choi, P.Y.; Cummings, J.A.; Cam, J.; Hu, Q.; Lake, T.; Pan, W.; Kastin, A.J.; et al. The Amazon rain forest plant Uncaria tomentosa (cat’s claw) and its specific proanthocyanidin constituents are potent inhibitors and reducers of both brain plaques and tangles. Sci. Rep. 2019, 9, 561. [Google Scholar] [CrossRef] [Green Version]
  60. Huang, M.; Jiang, X.; Liang, Y.; Liu, Q.; Chen, S.; Guo, Y. Berberine improves cognitive impairment by promoting autophagic clearance and inhibiting production of β-amyloid in APP/tau/PS1 mouse model of Alzheimer’s disease. Exp. Gerontol. 2017, 91, 25–33. [Google Scholar] [CrossRef] [PubMed]
  61. Mani, R.J.; Mittal, K.; Katare, D.P. Protective Effects of Quercetin in Zebrafish Model of Alzheimer’s Disease. Asian J. Pharm. 2018, 12, S660. [Google Scholar]
  62. Leiteritz, A.; Dilberger, B.; Wenzel, U.; Fitzenberger, E. Betaine reduces β-amyloid-induced paralysis through activation of cystathionine-β-synthase in an Alzheimer model of Caenorhabditis elegans. Genes Nutr. 2018, 13, 21. [Google Scholar] [CrossRef] [Green Version]
  63. Finley, J.W.; Gao, S. A Perspective on Crocus sativus L. (Saffron) Constituent Crocin: A Potent Water-Soluble Antioxidant and Potential Therapy for Alzheimer’s Disease. J. Agric. Food Chem. 2017, 65, 1005–1020. [Google Scholar] [CrossRef] [PubMed]
  64. Singh, M.; Ramassamy, C. In vitro screening of neuroprotective activity of Indian medicinal plant Withania somnifera. J. Nutr. Sci. 2017, 6, e54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kim, J.K.; Bae, H.; Kim, M.-J.; Choi, S.J.; Cho, H.Y.; Hwang, H.-J.; Kim, Y.J.; Lim, S.T.; Kim, E.K.; Kim, H.K.; et al. Inhibitory Effect of Poncirus trifoliate on Acetylcholinesterase and Attenuating Activity against Trimethyltin-Induced Learning and Memory Impairment. Biosci. Biotechnol. Biochem. 2009, 73, 1105–1112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Bihaqi, S.W.; Sharma, M.; Singh, A.P.; Tiwari, M. Neuroprotective role of Convolvulus pluricaulis on aluminium induced neurotoxicity in rat brain. J. Ethnopharmacol. 2009, 124, 409–415. [Google Scholar] [CrossRef]
  67. Azimi, A.; Ghaffari, S.M.; Riazi, G.H.; Arab, S.S.; Tavakol, M.M.; Pooyan, S. α-Cyperone of Cyperus rotundus is an effective candidate for reduction of inflammation by destabilization of microtubule fibers in brain. J. Ethnopharmacol. 2016, 194, 219–227. [Google Scholar] [CrossRef]
  68. Rivera, D.S.; Lindsay, C.; Codocedo, J.F.; Morel, I.; Pinto, C.; Cisternas, P.; Bozinovic, F.; Inestrosa, N.C. Andrographolide recovers cognitive impairment in a natural model of Alzheimer’s disease (Octodon degus). Neurobiol. Aging 2016, 46, 204–220. [Google Scholar] [CrossRef] [PubMed]
  69. Balez, R.; Steiner, N.; Engel, M.; Muñoz, S.S.; Lum, J.S.; Wu, Y.; Wang, D.; Vallotton, P.; Sachdev, P.; O’connor, M.; et al. Neuroprotective effects of apigenin against inflammation, neuronal excitability and apoptosis in an induced pluripotent stem cell model of Alzheimer’s disease. Sci. Rep. 2016, 6, 31450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Shi, J.; Li, Y.; Zhang, Y.; Chen, J.; Gao, J.; Zhang, T.; Shang, X.; Zhang, X. Baicalein Ameliorates Aβ-Induced Memory Deficits and Neuronal Atrophy via Inhibition of PDE2 and PDE4. Front. Pharmacol. 2021, 12, 794458. [Google Scholar] [CrossRef]
  71. Celik Topkara, K.; Kilinc, E.; Cetinkaya, A.; Saylan, A.; Demir, S. Therapeutic effects of carvacrol on beta-amyloid-induced impairments in in vitro and in vivo models of Alzheimer’s disease. Eur. J. Neurosci. 2022, 56, 5714–5726. [Google Scholar] [CrossRef] [PubMed]
  72. Li, L.; Li, W.; Jung, S.W.; Lee, Y.W.; Kim, Y.H. Protective Effects of Decursin and Decursinol Angelate against Amyloid β-Protein-Induced Oxidative Stress in the PC12 Cell Line: The Role of Nrf2 and Antioxidant Enzymes. Biosci. Biotechnol. Biochem. 2011, 75, 434–442. [Google Scholar] [CrossRef] [Green Version]
  73. Duan, X.; Li, Y.; Xu, F.; Ding, H. Study on the neuroprotective effects of Genistein on Alzheimer’s disease. Brain Behav. 2021, 11, e02100. [Google Scholar] [CrossRef] [PubMed]
  74. Huang, D.S.; Yu, Y.C.; Wu, C.H.; Lin, J.Y. Protective Effects of Wogonin against Alzheimer’s Disease by Inhibition of Amyloidogenic Pathway. Evid.-Based Complement. Altern. Med. 2017, 2017, 3545169. [Google Scholar] [CrossRef] [Green Version]
  75. Xu, P.-X.; Wang, S.-W.; Yu, X.-L.; Su, Y.-J.; Wang, T.; Zhou, W.-W.; Zhang, H.; Wang, Y.-J.; Liu, R.-T. Rutin improves spatial memory in Alzheimer’s disease transgenic mice by reducing Aβ oligomer level and attenuating oxidative stress and neuroinflammation. Behav. Brain Res. 2014, 264, 173–180. [Google Scholar] [CrossRef] [PubMed]
  76. Daily, J.W.; Kang, S.; Park, S. Protection against Alzheimer’s disease by luteolin: Role of brain glucose regulation, anti-inflammatory activity, and the gut microbiota-liver-brain axis. BioFactors 2021, 47, 218–231. [Google Scholar] [CrossRef] [PubMed]
  77. Sabogal-Guáqueta, A.M.; Osorio, E.; Cardona-Gómez, G.P. Linalool reverses neuropathological and behavioral impairments in old triple transgenic Alzheimer’s mice. Neuropharmacology 2016, 102, 111–120. [Google Scholar] [CrossRef] [Green Version]
  78. Asiatic Acid Nullified Aluminium Toxicity in In Vitro Model of Alzheimer’s Disease. Available online: https://www.imrpress.com/journal/FBE/10/2/10.2741/E823 (accessed on 30 May 2023).
  79. Li, Z.; Zhao, T.; Li, J.; Yu, Q.; Feng, Y.; Xie, Y.; Sun, P. Nanomedicine Based on Natural Products: Improving Clinical Application Potential. J. Nanomater. 2022, 2022, 3066613. [Google Scholar] [CrossRef]
  80. Woon, C.K.; Hui, W.K.; Abas, R.; Haron, M.H.; Das, S.; Lin, T.S. Natural Product-based Nanomedicine: Recent Advances and Issues for the Treatment of Alzheimer’s Disease. Curr. Neuropharmacol. 2022, 20, 1498–1518. [Google Scholar] [CrossRef]
  81. Yu, A.M.; Jian, C.; Yu, A.H.; Tu, M.J. RNA therapy: Are we using the right molecules? Pharmacol. Ther. 2019, 196, 91–104. [Google Scholar] [CrossRef]
  82. Shin, H.; Park, S.-J.; Yim, Y.; Kim, J.; Choi, C.; Won, C.; Min, D.-H. Recent Advances in RNA Therapeutics and RNA Delivery Systems Based on Nanoparticles. Adv. Ther. 2018, 1, 1800065. [Google Scholar] [CrossRef]
  83. Zogg, H.; Singh, R.; Ro, S. Current Advances in RNA Therapeutics for Human Diseases. Int. J. Mol. Sci. 2022, 23, 2736. [Google Scholar] [CrossRef] [PubMed]
  84. Mollocana-Lara, E.C.; Ni, M.; Agathos, S.N.; Gonzales-Zubiate, F.A. The infinite possibilities of RNA therapeutics. J. Ind. Microbiol. Biotechnol. 2021, 48, kuab063. [Google Scholar] [CrossRef]
  85. DeLong, R. Ushering in a new era of RNA-based therapies. Commun. Biol. 2021, 4, 577. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, Y.K. RNA therapy: Rich history, various applications and unlimited future prospects. Exp. Mol. Med. 2022, 54, 455–465. [Google Scholar] [CrossRef] [PubMed]
  87. Kim, Y.K. RNA Therapy: Current Status and Future Potential. Chonnam Med. J. 2020, 56, 87. [Google Scholar] [CrossRef] [PubMed]
  88. Anthony, K. RNA-based therapeutics for neurological diseases. RNA Biol. 2022, 19, 176–190. [Google Scholar] [CrossRef]
  89. Lee, M.J.; Lee, I.; Wang, K. Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders. Biomedicines 2022, 10, 158. [Google Scholar] [CrossRef] [PubMed]
  90. Jurcău, M.C.; Andronie-Cioara, F.L.; Jurcău, A.; Marcu, F.; Ţiț, D.M.; Pașcalău, N.; Nistor-Cseppentö, D.C. The Link between Oxidative Stress, Mitochondrial Dysfunction and Neuroinflammation in the Pathophysiology of Alzheimer’s Disease: Therapeutic Implications and Future Perspectives. Antioxidants 2022, 11, 2167. [Google Scholar] [CrossRef] [PubMed]
  91. Walgrave, H.; Zhou, L.; De Strooper, B.; Salta, E. The promise of microRNA-based therapies in Alzheimer’s disease: Challenges and perspectives. Mol. Neurodegener. 2021, 16, 76. [Google Scholar] [CrossRef] [PubMed]
  92. Kreth, S.; Hübner, M.; Hinske, L.C. MicroRNAs as Clinical Biomarkers and Therapeutic Tools in Perioperative Medicine. Obstet. Anesth. Dig. 2018, 126, 670–681. [Google Scholar] [CrossRef]
  93. Martier, R.; Konstantinova, P. Gene Therapy for Neurodegenerative Diseases: Slowing down the Ticking Clock. Front. Neurosci. 2020, 14, 580179. [Google Scholar] [CrossRef] [PubMed]
  94. Zhou, Y.; Zhu, F.; Liu, Y.; Zheng, M.; Wang, Y.; Zhang, D.; Anraku, Y.; Zou, Y.; Li, J.; Wu, H.; et al. Blood-brain barrier–penetrating siRNA nanomedicine for Alzheimer’s disease therapy. Sci. Adv. 2020, 6, eabc7031. [Google Scholar] [CrossRef] [PubMed]
  95. Long, J.M.; Lahiri, D.K. MicroRNA-101 downregulates Alzheimer’s amyloid-β precursor protein levels in human cell cultures and is differentially expressed. Biochem. Biophys. Res. Commun. 2011, 404, 889–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Liu, W.; Zhao, J.; Lu, G. miR-106b inhibits tau phosphorylation at Tyr18 by targeting Fyn in a model of Alzheimer’s disease. Biochem. Biophys. Res. Commun. 2016, 478, 852–857. [Google Scholar] [CrossRef] [PubMed]
  97. Jiang, Y.; Xu, B.; Chen, J.; Sui, Y.; Ren, L.; Li, J.; Zhang, H.; Guo, L.; Sun, X. Micro-RNA-137 Inhibits Tau Hyperphosphorylation in Alzheimer’s Disease and Targets the CACNA1C Gene in Transgenic Mice and Human Neuroblastoma SH-SY5Y Cells. Med. Sci. Monit. 2018, 24, 5635–5644. [Google Scholar] [CrossRef]
  98. Santa-Maria, I.; Alaniz, M.E.; Renwick, N.; Cela, C.; Fulga, T.A.; Van Vactor, D.; Tuschl, T.; Clark, L.N.; Shelanski, M.L.; McCabe, B.D.; et al. Dysregulation of microRNA-219 promotes neurodegeneration through post-transcriptional regulation of tau. J. Clin. Investig. 2015, 125, 681–686. [Google Scholar] [CrossRef] [Green Version]
  99. Estfanous, S.; Daily, K.P.; Eltobgy, M.; Deems, N.P.; Anne, M.N.K.; Krause, K.; Badr, A.; Hamilton, K.; Carafice, C.; Hegazi, A.; et al. Elevated Expression of MiR-17 in Microglia of Alzheimer’s Disease Patients Abrogates Autophagy-Mediated Amyloid-β Degradation. Front. Immunol. 2021, 12, 705581. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, R.; Chopra, N.; Nho, K.; Maloney, B.; Obukhov, A.G.; Nelson, P.T.; Counts, S.E.; Lahiri, D.K. Human microRNA (miR-20b-5p) modulates Alzheimer’s disease pathways and neuronal function, and a specific polymorphism close to the MIR20B gene influences Alzheimer’s biomarkers. Mol. Psychiatry 2022, 27, 1256–1273. [Google Scholar] [CrossRef]
  101. Zong, Y.; Wang, H.; Dong, W.; Quan, X.; Zhu, H.; Xu, Y.; Huang, L.; Ma, C.; Qin, C. miR-29c regulates BACE1 protein expression. Brain Res. 2011, 1395, 108–115. [Google Scholar] [CrossRef]
  102. Chopra, N.; Wang, R.; Maloney, B.; Nho, K.; Beck, J.S.; Pourshafie, N.; Niculescu, A.; Saykin, A.J.; Rinaldi, C.; Counts, S.E.; et al. MicroRNA-298 reduces levels of human amyloid-β precursor protein (APP), β-site APP-converting enzyme 1 (BACE1) and specific tau protein moieties. Mol. Psychiatry 2021, 26, 5636–5657. [Google Scholar] [CrossRef] [PubMed]
  103. He, C.; Su, C.; Zhang, W.; Wan, Q. miR-485-5p alleviates Alzheimer’s disease progression by targeting PACS1. Transl. Neurosci. 2021, 12, 335–345. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, J.; Zuo, X.; Han, J.; Dai, Q.; Xu, H.; Liu, Y.; Cui, S. MiR-9-5p inhibits mitochondrial damage and oxidative stress in AD cell models by targeting GSK-3β. Biosci. Biotechnol. Biochem. 2020, 84, 2273–2280. [Google Scholar] [CrossRef] [PubMed]
  105. Deng, Y.; Zhang, J.; Sun, X.; Ma, G.; Luo, G.; Miao, Z.; Song, L. miR-132 improves the cognitive function of rats with Alzheimer’s disease by inhibiting the MAPK1 signal pathway. Exp. Ther. Med. 2020, 20, 159. [Google Scholar] [CrossRef]
  106. Liang, C.; Zhu, H.; Xu, Y.; Huang, L.; Ma, C.; Deng, W.; Liu, Y.; Qin, C. MicroRNA-153 negatively regulates the expression of amyloid precursor protein and amyloid precursor-like protein 2. Brain Res. 2012, 1455, 103–113. [Google Scholar] [CrossRef] [PubMed]
  107. Xu, H.; Rosler, T.W.; Carlsson, T.; Andrade, A.; Fiala, O.; Hollerhage, M.; Oertel, W.H.; Goedert, M.; Aigner, A.; Hoglinger, G.U. Tau Silencing by siRNA in the P301S Mouse Model of Tauopathy. Curr. Gene Ther. 2014, 14, 343–351. [Google Scholar] [CrossRef]
  108. Kandimalla, R.J.; Wani, W.Y.; Bk, B.; Gill, K.D. siRNA against presenilin 1 (PS1) down regulates amyloid b42 production in IMR-32 cells. J. Biomed. Sci. 2012, 19, 2. [Google Scholar] [PubMed] [Green Version]
  109. Senechal, Y.; Kelly, P.H.; Cryan, J.F.; Natt, F.; Dev, K.K. Amyloid precursor protein knockdown by siRNA impairs spontaneous alternation in adult mice: In vivo knockdown of APP by RNAi. J. Neurochem. 2007, 102, 1928–1940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Lin, C.Y.; Perche, F.; Ikegami, M.; Uchida, S.; Kataoka, K.; Itaka, K. Messenger RNA-based therapeutics for brain diseases: An animal study for augmenting clearance of beta-amyloid by intracerebral administration of neprilysin mRNA loaded in polyplex nanomicelles. J. Control. Release 2016, 235, 268–275. [Google Scholar] [CrossRef]
  111. Lim, S.A.; Cox, A.; Tung, M.; Chung, E.J. Clinical progress of nanomedicine-based RNA therapies. Bioact. Mater. 2022, 12, 203–213. [Google Scholar] [CrossRef] [PubMed]
  112. Gorshkov, A.; Purvinsh, L.; Brodskaia, A.; Vasin, A. Exosomes as Natural Nanocarriers for RNA-Based Therapy and Prophylaxis. Nanomaterials 2022, 12, 524. [Google Scholar] [CrossRef] [PubMed]
  113. Tsakiri, M.; Zivko, C.; Demetzos, C.; Mahairaki, V. Lipid-based nanoparticles and RNA as innovative neuro-therapeutics. Front. Pharmacol. 2022, 13, 900610. [Google Scholar] [CrossRef] [PubMed]
  114. Fernandes, F.; Dias-Teixeira, M.; Delerue-Matos, C.; Grosso, C. Critical Review of Lipid-Based Nanoparticles as Carriers of Neuroprotective Drugs and Extracts. Nanomaterials 2021, 11, 563. [Google Scholar] [CrossRef] [PubMed]
  115. Pardridge, W.M. Treatment of Alzheimer’s Disease and Blood–Brain Barrier Drug Delivery. Pharmaceuticals 2020, 13, 394. [Google Scholar] [CrossRef]
  116. Wohlfart, S.; Gelperina, S.; Kreuter, J. Transport of drugs across the blood–brain barrier by nanoparticles. J. Control. Release 2012, 161, 264–273. [Google Scholar] [CrossRef] [PubMed]
  117. Zhou, Y.; Peng, Z.; Seven, E.S.; Leblanc, R.M. Crossing the blood-brain barrier with nanoparticles. J. Control. Release 2018, 270, 290–303. [Google Scholar] [CrossRef] [PubMed]
  118. Bors, L.; Erdő, F. Overcoming the Blood–Brain Barrier. Challenges and Tricks for CNS Drug Delivery. Sci. Pharm. 2019, 87, 6. [Google Scholar] [CrossRef] [Green Version]
  119. Wong, K.H.; Riaz, M.K.; Xie, Y.; Zhang, X.; Liu, Q.; Chen, H.; Bian, Z.; Chen, X.; Lu, A.; Yang, Z. Review of Current Strategies for Delivering Alzheimer’s Disease Drugs across the Blood-Brain Barrier. Int. J. Mol. Sci. 2019, 20, 381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Zenaro, E.; Piacentino, G.; Constantin, G. The blood-brain barrier in Alzheimer’s disease. Neurobiol. Dis. 2017, 107, 41–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Sharma, C.; Woo, H.; Kim, S.R. Addressing Blood–Brain Barrier Impairment in Alzheimer’s Disease. Biomedicines 2022, 10, 742. [Google Scholar] [CrossRef] [PubMed]
  122. Juhairiyah, F.; de Lange, E.C.M. Understanding Drug Delivery to the Brain Using Liposome-Based Strategies: Studies that Provide Mechanistic Insights Are Essential. AAPS J. 2021, 23, 114. [Google Scholar] [CrossRef] [PubMed]
  123. Satapathy, M.K.; Yen, T.-L.; Jan, J.-S.; Tang, R.-D.; Wang, J.-Y.; Taliyan, R.; Yang, C.-H. Solid Lipid Nanoparticles (SLNs): An Advanced Drug Delivery System Targeting Brain through BBB. Pharmaceutics 2021, 13, 1183. [Google Scholar] [CrossRef] [PubMed]
  124. Musielak, E.; Feliczak-Guzik, A.; Nowak, I. Synthesis and Potential Applications of Lipid Nanoparticles in Medicine. Materials 2022, 15, 682. [Google Scholar] [CrossRef]
  125. Heidarzadeh, M.; Gürsoy-Özdemir, Y.; Kaya, M.; Abriz, A.E.; Zarebkohan, A.; Rahbarghazi, R.; Sokullu, E. Exosomal delivery of therapeutic modulators through the blood–brain barrier; promise and pitfalls. Cell Biosci. 2021, 11, 142. [Google Scholar] [CrossRef]
  126. Negahdaripour, M.; Vakili, B.; Nezafat, N. Exosome-based vaccines and their position in next generation vaccines. Int. Immunopharmacol. 2022, 113, 109265. [Google Scholar] [CrossRef] [PubMed]
  127. Jafari, D.; Malih, S.; Eini, M.; Jafari, R.; Gholipourmalekabadi, M.; Sadeghizadeh, M.; Samadikuchaksaraei, A. Improvement, scaling-up, and downstream analysis of exosome production. Crit. Rev. Biotechnol. 2020, 40, 1098–1112. [Google Scholar] [CrossRef] [PubMed]
  128. Hu, Q.; Su, H.; Li, J.; Lyon, C.; Tang, W.; Wan, M.; Hu, T.Y. Clinical applications of exosome membrane proteins. Precis. Clin. Med. 2020, 3, 54–66. [Google Scholar] [CrossRef] [PubMed]
  129. Ludwig, N.; Whiteside, T.L.; Reichert, T.E. Challenges in Exosome Isolation and Analysis in Health and Disease. Int. J. Mol. Sci. 2019, 20, 4684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Rashed, M.H.; Bayraktar, E.; Helal, G.K.; Abd-Ellah, M.F.; Amero, P.; Chavez-Reyes, A.; Rodriguez-Aguayo, C. Exosomes: From Garbage Bins to Promising Therapeutic Targets. Int. J. Mol. Sci. 2017, 18, 538. [Google Scholar] [CrossRef] [Green Version]
  131. Zhang, N.; He, F.; Li, T.; Chen, J.; Jiang, L.; Ouyang, X.-P.; Zuo, L. Role of Exosomes in Brain Diseases. Front. Cell Neurosci. 2021, 15, 743353. [Google Scholar] [CrossRef] [PubMed]
  132. Xu, L.; Wang, X.; Liu, Y.; Yang, G.; Falconer, R.J.; Zhao, C.X. Lipid Nanoparticles for Drug Delivery. Adv. NanoBiomed Res. 2022, 2, 2100109. [Google Scholar] [CrossRef]
  133. Feng, R.; Patil, S.; Zhao, X.; Miao, Z.; Qian, A. RNA Therapeutics—Research and Clinical Advancements. Front. Mol. Biosci. 2021, 8, 710738. [Google Scholar] [CrossRef]
  134. Burdușel, A.C.; Andronescu, E. Lipid Nanoparticles and Liposomes for Bone Diseases Treatment. Biomedicines 2022, 10, 3158. [Google Scholar] [CrossRef]
  135. Duan, Y.; Dhar, A.; Patel, C.; Khimani, M.; Neogi, S.; Sharma, P.; Kumar, N.S.; Vekariya, R.L. A brief review on solid lipid nanoparticles: Part and parcel of contemporary drug delivery systems. RSC Adv. 2020, 10, 26777–26791. [Google Scholar] [CrossRef] [PubMed]
  136. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles—From Liposomes to mRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef]
  137. Abdellatif, A.A.; Younis, M.A.; Alsowinea, A.F.; Abdallah, E.M.; Abdel-Bakky, M.S.; Al-Subaiyel, A.; Hassan, Y.A.; Tawfeek, H.M. Lipid nanoparticles technology in vaccines: Shaping the future of prophylactic medicine. Colloids Surf. B Biointerfaces 2023, 222, 113111. [Google Scholar] [CrossRef] [PubMed]
  138. Vieira, D.; Gamarra, L. Getting into the brain: Liposome-based strategies for effective drug delivery across the blood–brain barrier. Int. J. Nanomed. 2016, 11, 5381–5414. [Google Scholar] [CrossRef] [Green Version]
  139. Gauro, R.; Nandave, M.; Jain, V.K.; Jain, K. Advances in dendrimer-mediated targeted drug delivery to the brain. J. Nanoparticle Res. 2021, 23, 76. [Google Scholar] [CrossRef]
  140. Teleanu, D.; Chircov, C.; Grumezescu, A.; Volceanov, A.; Teleanu, R. Blood-Brain Delivery Methods Using Nanotechnology. Pharmaceutics 2018, 10, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Sela, H.; Cohen, H.; Elia, P.; Zach, R.; Karpas, Z.; Zeiri, Y. Spontaneous penetration of gold nanoparticles through the blood brain barrier (BBB). J. Nanobiotechnol. 2015, 13, 71. [Google Scholar] [CrossRef] [Green Version]
  142. Li, X.; Corbett, A.L.; Taatizadeh, E.; Tasnim, N.; Little, J.P.; Garnis, C.; Daugaard, M.; Guns, E.; Hoorfar, M.; Li, I.T.S. Challenges and opportunities in exosome research—Perspectives from biology, engineering, and cancer therapy. APL Bioeng. 2019, 3, 011503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Schiffelers, R.; Kooijmans, S.; Vader, P.; Dommelen, V.; Solinge, V. Exosome mimetics: A novel class of drug delivery systems. Int. J. Nanomed. 2012, 7, 1525–1541. [Google Scholar] [CrossRef] [Green Version]
  144. Fernandes, M.; Lopes, I.; Magalhães, L.; Sárria, M.P.; Machado, R.; Sousa, J.C.; Botelho, C.; Teixeira, J.; Gomes, A.C. Novel concept of exosome-like liposomes for the treatment of Alzheimer’s disease. J. Control. Release 2021, 336, 130–143. [Google Scholar] [CrossRef]
  145. Zha, Y.; Lin, T.; Li, Y.; Zhang, X.; Wang, Z.; Li, Z.; Ye, Y.; Wang, B.; Zhang, S.; Wang, J. Exosome-mimetics as an engineered gene-activated matrix induces in-situ vascularized osteogenesis. Biomaterials 2020, 247, 119985. [Google Scholar] [CrossRef] [PubMed]
  146. Severic, M.; Ma, G.; Pereira, S.G.T.; Ruiz, A.; Cheung, C.C.; Al-Jamal, W.T. Genetically-engineered anti-PSMA exosome mimetics targeting advanced prostate cancer in vitro and in vivo. J. Control. Release 2021, 330, 101–110. [Google Scholar] [CrossRef]
  147. Lee, C.-S.; Fan, J.; Hwang, H.S.; Kim, S.; Chen, C.; Kang, M.; Aghaloo, T.; James, A.W.; Lee, M. Bone-Targeting Exosome Mimetics Engineered by Bioorthogonal Surface Functionalization for Bone Tissue Engineering. Nano Lett. 2023, 23, 1202–1210. [Google Scholar] [CrossRef] [PubMed]
  148. Fan, J.; Lee, C.-S.; Kim, S.; Chen, C.; Aghaloo, T.; Lee, M. Generation of Small RNA-Modulated Exosome Mimetics for Bone Regeneration. ACS Nano 2020, 14, 11973–11984. [Google Scholar] [CrossRef] [PubMed]
  149. Wang, J.; Li, M.; Jin, L.; Guo, P.; Zhang, Z.; Zhanghuang, C.; Tan, X.; Mi, T.; Liu, J.; Wu, X.; et al. Exosome mimetics derived from bone marrow mesenchymal stem cells deliver doxorubicin to osteosarcoma in vitro and in vivo. Drug Deliv. 2022, 29, 3291–3303. [Google Scholar] [CrossRef]
  150. Jang, S.C.; Kim, O.Y.; Yoon, C.M.; Choi, D.-S.; Roh, T.-Y.; Park, J.; Nilsson, J.; Lötvall, J.; Kim, Y.-K.; Gho, Y.S. Bioinspired Exosome-Mimetic Nanovesicles for Targeted Delivery of Chemotherapeutics to Malignant Tumors. ACS Nano 2013, 7, 7698–7710. [Google Scholar] [CrossRef]
  151. Wu, J.-Y.; Li, Y.-J.; Wang, J.; Hu, X.-B.; Huang, S.; Luo, S.; Xiang, D.-X. Multifunctional exosome-mimetics for targeted anti-glioblastoma therapy by manipulating protein corona. J. Nanobiotechnol. 2021, 19, 405. [Google Scholar] [CrossRef]
  152. Yan, Y.; Du, C.; Duan, X.; Yao, X.; Wan, J.; Jiang, Z.; Qin, Z.; Li, W.; Pan, L.; Gu, Z.; et al. Inhibiting collagen I production and tumor cell colonization in the lung via miR-29a-3p loading of exosome-/liposome-based nanovesicles. Acta Pharm. Sin. B 2022, 12, 939–951. [Google Scholar] [CrossRef]
Figure 1. Hallmarks of neurodegenerative diseases. The scheme illustrates the six hallmarks of these disorders: Protein aggregation, disruptive proteostasis, neuroinflammation, oxidative stress, synaptic failure, and neuronal death. Created in BioRender.com (accessed on 25 May 2023).
Figure 1. Hallmarks of neurodegenerative diseases. The scheme illustrates the six hallmarks of these disorders: Protein aggregation, disruptive proteostasis, neuroinflammation, oxidative stress, synaptic failure, and neuronal death. Created in BioRender.com (accessed on 25 May 2023).
Molecules 28 06015 g001
Figure 2. Alzheimer’s disease hallmarks. (A) Pathway that leads to the formation of amyloid plaques and the mechanism responsible for the formation of neurofibrillary tangles. (B) Accumulation of amyloid plaques and neurofibrillary tangles provoke disorders such as synaptic dysfunction, neuroinflammation, oxidative stress, and higher activity of acetylcholinesterase. These result in neurodegeneration. Created in BioRender.com (accessed on 20 April 2023).
Figure 2. Alzheimer’s disease hallmarks. (A) Pathway that leads to the formation of amyloid plaques and the mechanism responsible for the formation of neurofibrillary tangles. (B) Accumulation of amyloid plaques and neurofibrillary tangles provoke disorders such as synaptic dysfunction, neuroinflammation, oxidative stress, and higher activity of acetylcholinesterase. These result in neurodegeneration. Created in BioRender.com (accessed on 20 April 2023).
Molecules 28 06015 g002
Figure 3. Potential problems of natural products in vivo and advantages of natural products delivered by nanoparticles. Created in BioRender.com (accessed on 5 May 2023).
Figure 3. Potential problems of natural products in vivo and advantages of natural products delivered by nanoparticles. Created in BioRender.com (accessed on 5 May 2023).
Molecules 28 06015 g003
Figure 4. Possible RNA-based therapeutic strategies for AD. (A) mRNA leads to protein expression and (B) siRNA, miRNA, and antisense oligonucleotide inhibit protein expression. Created in BioRender.com (accessed on 20 April 2023).
Figure 4. Possible RNA-based therapeutic strategies for AD. (A) mRNA leads to protein expression and (B) siRNA, miRNA, and antisense oligonucleotide inhibit protein expression. Created in BioRender.com (accessed on 20 April 2023).
Molecules 28 06015 g004
Figure 5. How RNA delivery systems can overcome the physiological obstacles faced by naked RNAs. (a) RNA can be delivered across the BBB; (b) protection of RNAs from degradation by ribonucleases (RNAses); (c) enhanced cellular internalization by controlled or specific pathways; (d) intracellular endosomal degradation can be avoided by endosomal escape; (e) improved RNA-mediated bioactivities; (f) enhanced accumulation of RNA in target tissue. Created in BioRender.com (accessed on 20 April 2023).
Figure 5. How RNA delivery systems can overcome the physiological obstacles faced by naked RNAs. (a) RNA can be delivered across the BBB; (b) protection of RNAs from degradation by ribonucleases (RNAses); (c) enhanced cellular internalization by controlled or specific pathways; (d) intracellular endosomal degradation can be avoided by endosomal escape; (e) improved RNA-mediated bioactivities; (f) enhanced accumulation of RNA in target tissue. Created in BioRender.com (accessed on 20 April 2023).
Molecules 28 06015 g005
Figure 6. Structural representation of the BBB anatomy (A) and possible pathways of entrance into the central nervous system (B). Created in BioRender.com (accessed on 20 April 2023).
Figure 6. Structural representation of the BBB anatomy (A) and possible pathways of entrance into the central nervous system (B). Created in BioRender.com (accessed on 20 April 2023).
Molecules 28 06015 g006
Figure 7. Exosome-like liposomes combine advantages of liposomes and exosomes, with the capacity to be loaded with different types of molecules due to their constitution. Created in BioRender.com (accessed on 20 April 2023).
Figure 7. Exosome-like liposomes combine advantages of liposomes and exosomes, with the capacity to be loaded with different types of molecules due to their constitution. Created in BioRender.com (accessed on 20 April 2023).
Molecules 28 06015 g007
Table 1. Natural compounds or extracts that can be used for AD therapy.
Table 1. Natural compounds or extracts that can be used for AD therapy.
Natural CompoundRole in ADReferences
EugenolRats were fed aluminum, a neurotoxic metal that leads to oxidative brain injury and enhanced lipid peroxidation, disruption of neurotrophic, cholinergic, and serotonergic functions, and induce apoptosis with ultimate neuronal and astrocyte damages. A neuroprotective role of eugenol against the aluminum effects was verified through its antioxidant, antiapoptotic potential and its neurotrophic properties.[51]
MentholMenthol inhalation by mice (1 week per month, for 6 months) prevented cognitive impairment in the APP/PS1 mouse model of Alzheimer’s. [52]
ChrysinChrysin showed the ability to act as a membrane shield against early oxidative events mediated by O2˙- and other ROS that contribute to neuronal death triggered by AlCl3 exposure, showing chrysin’s neuroprotective action.[53]
Rosmarinic acidSuppresses Aβ accumulation in mice.[54]
Ginkgo bilobaGinkgo biloba improves microcirculation, inhibits the expression of inflammatory factors, and reduces inflammatory damage to neurons, thereby improving the spatial exploration memory of dementia model rats.[55]
ResveratrolMultiple studies demonstrated that resveratrol has neuroprotective, anti-inflammatory, and antioxidant characteristics and the ability to minimize Aβ peptide aggregation and toxicity in the hippocampus of Alzheimer’s patients, stimulating neurogenesis and inhibiting hippocampal degeneration. Furthermore, resveratrol’s antioxidant effect promotes neuronal development by activating the silent information regulator-1, which can protect against the detrimental effects of oxidative stress.[56]
Huperzine AHuperzine A is natural, potent, highly specific reversible inhibitor of acetylcholinesterase, with the ability to cross the BBB.[57]
BrahmiThe neuroprotective properties of Brahmi include the reduction of ROS and neuroinflammation, the inhibition of the aggregation of Aβ and the improvement of cognitive and learning behavior.[58]
Uncaria tomentosaInhibits plaques and tangles formation.[59]
BerberineBerberine has antioxidant activity and promotes AChE and monoamine oxidase inhibition. Berberine has been shown to improve memory, lower Aβ and APP concentration, and diminish Aβ plaque accumulation.[60]
QuercetinBehavioral and biochemical tests confirm that quercetin promotes the reduction in oxidative stress and increased cognition in zebrafish AD models induced with aluminum chloride.[61]
BetaineBetaine has been shown to decrease homocysteine levels and Aβ toxicity in Caenorhabditis elegans AD model.[62]
CurcuminCurcumin is known to be a potent antioxidant, anti-inflammatory and anti-amyloidogenic compound, that plays a beneficial role in treating AD through several mechanisms. Curcumin can promote a significant reduction of Aβ oligomers and fibril formation.[46]
CrocinCrocin, the main constituent of Crocus sativus L., has a multifunctional role in protecting brain cells, modulating aggregation of Aβ and Tau proteins, attenuating cognitive and memory impairments, and improving oxidative stress.[63]
Withania somniferaWithania somnifera extract can protect against Aβ peptide- and acrolein-induced toxicity. Treatment with this extract significantly protected against Aβ and acrolein, in various cell survival assays with the human neuroblastoma cell line SK-N-SH, significantly reduced the generation of ROS and was demonstrated to be a potent inhibitor of AChE activity.[64]
Poncirus trifoliateThe extract of Poncirus trifoliate is a naturally occurring AChE inhibitor. It showed a 47.31% inhibitory effect on the activity of acetylcholine.[65]
Convolvulus pluricaulisConvolvulus pluricaulis prevented aluminum-induced neurotoxicity in rat cerebral cortex. [66]
α-Cyperoneα-Cyperone binds and interacts with tubulin, being capable of destabilizing microtubule polymerization. The effect of this interaction could result in reduction of inflammation.[67]
AndrographolideAndrographolide has beneficial effects in the recovery of spatial memory and learning performance, recovery of synaptic basal transmission, partial or complete protection of certain synaptic proteins and shows a specific neuroprotective effect, that includes the reduction of phosphorylated Tau and Aβ aggregate maturation, in aged degus. [68]
ApigeninApigenin has been shown to have anti-inflammatory and neuroprotective properties in a number of cell and animal models. This compound is also able to protect human induced pluripotent stem cell-derived AD neurons via multiple pathways, by reducing the frequency of spontaneous Ca2+ signals and significantly reducing caspase-3/7 mediated apoptosis.[69]
BaicaleinBaicalein has antioxidant and anti-inflammatory effects.[70]
CarvacrolCarvacrol possesses anti-AChE, antioxidant, and neuroprotective properties. This compound alleviated Aβ-induced deficits by reducing cellular neurotoxicity and oxidative stress in the SH-SY5Y cell line, and by reducing oxidative stress and memory impairment in a rat model of AD. [71]
Decursin/Decursinol angelateDecursin and decursinol angelate increase cellular resistance to Aβ-induced oxidative injury in PC12 cells.[72]
GenisteinIn vivo studies have shown that genistein improves brain function, antagonizes the toxicity of Aβ and has neuroprotective effects.[73]
WogoninWogonin has various neuroprotective and neurotrophic activities, such as inducing neurite outgrowth.[74]
RutinRutin is antioxidant, anti-inflammatory, and has the capacity of reducing Aβ oligomer activities.[75]
LuteolinLuteolin has the capacity to cross the BBB and can inhibit β- and γ-secretase to decrease Aβ. It can also reduce neuroinflammation and attenuate the phosphorylation of Tau. [76]
LinaloolA linalool-treated mice model of AD showed improved learning and spatial memory. This compound reverses the histopathological hallmarks of AD and restores cognitive and emotional functions via an anti-inflammatory effect.[77]
Asiatic acidPre-treatment with Asiatic Acid enhanced cell viability, attenuated rotenone-induced ROS, mitochondrial membrane dysfunction and apoptosis regulating AKT/GSK-3β signaling pathway, after aluminum maltolate neurotoxicity induction in SH-SY5Y neuroblastoma cells. [78]
Table 3. Examples of applications of exosome-like liposomes.
Table 3. Examples of applications of exosome-like liposomes.
Exosome-like Liposomes ApplicationsReferences
Exosome mimetics-mediated gene-activated matrix encapsulating the plasmid of vascular endothelial growth factor (VEGF) was able to sustainably deliver VEGF gene and significantly enhance the vascularized osteogenesis in vivo. [145]
PSMA-exosome mimetics showed increased cellular internalization in PSMA-positive PC cell lines (LNCaP and C4-2B) and higher tumor targeting was observed in solid C4-2B tumors, following intravenous administration, confirming their targeting ability in vivo.[146]
Exosome mimetics are reported for bone targeting involving the introduction of hydroxyapatite-binding moieties through bioorthogonal functionalization. Bone-binding ability of the engineered exosome mimetics is verified with hydroxyapatite-coated scaffolds and an ex vivo bone-binding assay.[147]
Administration of mesenchymal stem cells-exosome mimetics in conjunction with an injectable chitosan hydrogel into mouse nonhealing calvarial defects demonstrated robust bone regeneration. [148]
Bone marrow mesenchymal stem cells were sequentially extruded to generate exosome-mimetic to encapsulate doxorubicin to treat osteosarcoma. The results showed that demonstrated significantly more potent tumor inhibition activity and fewer side effects than free doxorubicin.[149]
In vitro, chemotherapeutic drug-loaded exosome-mimetics induced TNF-R-stimulated endothelial cell death in a dose-dependent manner. In vivo, experiments in mice showed that the chemotherapeutic drug-loaded exosome-mimetics traffic to tumor tissue and reduce tumor growth without the adverse effects observed with equipotent free drug.[150]
Multifunctional exosomes-mimetics decorated with angiopep-2 (Ang-EM) incorporating Docetaxel, for enhancing glioblastoma drug delivery by manipulating protein corona, Ang-EM showed enhanced BBB penetration ability and targeting ability to the gioblastoma. Ang-EM-mediated delivery increased the concentration of docetaxel in the tumor area.[151]
A designed lung-targeting liposomal nanovesicle carrying miR-29a-3p that mimics the exosomes, significantly down-regulated collagen I secretion by lung fibroblasts in vivo, thus alleviating the establishment of a pro-metastatic environment for circulating lung tumor cells.[152]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ribeiro, J.; Lopes, I.; Gomes, A.C. A New Perspective for the Treatment of Alzheimer’s Disease: Exosome-like Liposomes to Deliver Natural Compounds and RNA Therapies. Molecules 2023, 28, 6015. https://doi.org/10.3390/molecules28166015

AMA Style

Ribeiro J, Lopes I, Gomes AC. A New Perspective for the Treatment of Alzheimer’s Disease: Exosome-like Liposomes to Deliver Natural Compounds and RNA Therapies. Molecules. 2023; 28(16):6015. https://doi.org/10.3390/molecules28166015

Chicago/Turabian Style

Ribeiro, Joana, Ivo Lopes, and Andreia Castro Gomes. 2023. "A New Perspective for the Treatment of Alzheimer’s Disease: Exosome-like Liposomes to Deliver Natural Compounds and RNA Therapies" Molecules 28, no. 16: 6015. https://doi.org/10.3390/molecules28166015

Article Metrics

Back to TopTop