Next Article in Journal
Differential Roles of CD36 in Regulating Muscle Insulin Response Depend on Palmitic Acid Load
Previous Article in Journal
Identifying Patients in Whom the Follow-Up Scheme after Robot-Assisted Radical Prostatectomy Could Be Optimized in the First Year after Surgery: Reducing Healthcare Burden
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles, Cell-Penetrating Peptides and miRNAs as Future Novel Therapeutic Interventions for Parkinson’s and Alzheimer’s Disease

by
Cameron Noah Keighron
1,2,
Sahar Avazzadeh
1,2,
Katarzyna Goljanek-Whysall
1,3,
Brian McDonagh
1,
Linda Howard
4,
Thomas Ritter
4,5 and
Leo R. Quinlan
1,2,4,*
1
Physiology School of Medicine, University of Galway, H91 TK33 Galway, Ireland
2
Cellular Physiology Research Laboratory (CPRL), School of Medicine, University of Galway, H91 TK33 Galway, Ireland
3
Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK
4
Regenerative Medicine Institute, School of Medicine, College of Medicine, Nursing and Health Sciences, University of Galway, H91 TK33 Galway, Ireland
5
CURAM SFI Research Centre for Medical Devices, University of Galway, H91 TK33 Galway, Ireland
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(3), 728; https://doi.org/10.3390/biomedicines11030728
Submission received: 12 January 2023 / Revised: 16 February 2023 / Accepted: 25 February 2023 / Published: 28 February 2023

Abstract

:
Neurodegeneration is hallmarked by the progressive loss of dopaminergic neurons and/or a significant increase in protein aggregates in the brain. Neurodegenerative diseases are a leading cause of death worldwide with over 15 million people currently suffering from either Parkinson’s disease (PD) or Alzheimer’s disease (AD). PD is often characterized by both motor and non-motor symptoms, including muscle rigidity, tremors and bradykinesia, with AD displaying symptoms of confusion and dementia. The current mainstay of therapeutics includes pharmacological approaches such as levodopa to replace dopamine in PD patients, deep brain stimulation in affected regions of the brain and physical therapy. However, these treatments are typically not disease-modifying, though they do help at least for some time with symptom management. These treatments often also fail due to their inability to cross the blood–brain barrier. There is a need to develop new strategies to target neurodegeneration in an ever-ageing population. First, we review the current PD and AD treatments and their limitations. Second, we review the current use of extracellular vesicles (EVs), cell-penetrating peptides (CPPs) and miRNAs as neuroprotective agents. Finally, we discuss the possibility of exploiting these as a combinatory therapeutic, alongside some potential drawbacks.

1. Introduction

The hallmark of neurodegeneration is the progressive loss of brain function, often with overlapping biological and clinical symptoms [1]. In general, neurodegenerative diseases share common pathologies and pathways, most prominently protein aggregation, oxidative stress, neuroinflammation and blood–brain barrier dysfunction, leading primarily to the death of neurons from various regions of the brain [2,3,4]. Neuronal degeneration is profound in both Alzheimer’s (AD) and Parkinson’s disease (PD) with numerous overlapping pathways implicated, such as regional aggregation of intracellular proteins Tau and Alpha-synuclein [5]; complex genotype–phenotype relationships with common mutations such as leucine-rich repeat kinase 2 (LRRK2), PTEN-induced kinase 1 (PINK1) and amyloid precursor protein (APP) [6]; and alterations in pathways such as autophagy–lysosome activity, creating an imbalance between autophagosome formation and the autophagic degradation usually involved in clearing aggregated proteins [7]. Mitochondrial homeostasis is also implicated in AD and PD, leading to neuronal cell death via an increase in oxidative stress [8,9]. Lastly, innate immunity, synaptic toxicity and network dysfunction all contribute to the neuronal loss observed in AD and PD [10] (Figure 1).
AD and PD are the two most common neurodegenerative diseases worldwide. Cognitive dysfunction is the primary symptom exhibited in AD, while significant motor dysfunction is cardinal to PD [5]. AD is the most common cause of dementia, with a prevalence estimated at 24 million worldwide, which is expected to continue to rise over the next decade (GBD 2019 Dementia Forecasting Collaborators 2022). AD affects around 11% of the population over the age of 65, with PD affecting 2–3% of the population over 65 years of age, becoming the second most common neurodegenerative disease after AD [11]. AD can be classified into four stages: pre-clinical/pre-symptomatic, mild/early stage, moderate stage and severe/late stage [12,13,14]. These stages are often characterised by progressive memory loss, impaired balance, aphasia-like symptoms and an overall lack of independence in carrying out activities of daily living [15,16,17]. Two neuropathological changes have been identified in AD; positive lesions and negative lesions. Accumulation of neurofibrillary tangles, amyloid plaques and other deposits are significant indicators of AD, while on the other hand, neuronal, neuropil and synaptic loss-induced atrophy is also indicated in AD [18,19]. While the precise cause of the underlying pathological changes in AD is unknown, risk factors including age, genetics, traumatic brain injuries, diet and immune system dysregulation are some of the key contributors [20].
PD is associated with a lack of dopamine and an overall slowing of movement (bradykinesia) along with either a resting tremor or rigidity [21]. It has been suggested that there are two stages to PD, early (1 and 2) and late (3 and 4). In the early stages, symptoms can include rapid eye movement sleep behaviour disease (including sleep paralysis), as well as decreased smell, suggesting onset in the medulla and olfactory bulb. In stages 3 and 4, symptoms are more typical of cognitive impairment, including issues with movement and gait as well as hallucinations, suggesting that this stage’s pathology has progressed to the substantia nigra pars compacta and other midbrain and basal forebrain regions [22,23], often with aggregates of alpha-synuclein [23].

2. Aetiology and Pathophysiology of Disease

PD and AD are characterised by the presence of insoluble protein deposits, β-amyloid plaques and tau-containing neurofibrillary lesions in AD and α-synuclein-rich Lewy bodies in PD (Figure 1). Neuropathological changes in the disease progression and pathology of AD include neurofibrillary tangles, amyloid plaques, dystrophic neurites and neuropil threads [24]. PD presents with abnormal α-synuclein aggregates and the presence of Lewy bodies with selective loss of dopaminergic neurons [25,26]. Typically, PD is diagnosed at the mid to late stage of the disease due to a long period of dormancy between the initial loss of dopaminergic neurons and the development of more “typical” clinical symptoms [21,27]. AD follows a similar pathway with initial symptoms presenting over a 2–4-year period, then progressively worsening over the subsequent 10 years. PD and AD are largely considered to be sporadic, with evidence in recent years continuing to support the hypothesis that AD and PD may have substantial genetic components. In PD, this includes reported defects in the SNCA, PINK1 genes (associated with abnormal mitochondria and increased apoptosis), parkin (associated with impaired damaged protein tagging with ubiquitin), LRRK2 (associated with increased neuroinflammation) and DJ-1 (associated with increased reactive oxygen species), with genes such as APP (associated with the generation of beta-amyloid peptides), PSEN1 and PSEN2 (both interact with APP and are associated with the overproduction of toxic beta-amyloid peptides) displaying defects in AD [28,29,30,31].
Taking a closer look at both AD and PD suggests many commonalities in their pathophysiology, with many genes shared and co-expressed [32], as well as stark differences [33]. AD is well known for its increased concentration of Aβ42 (β-amyloid 42—due to mutations in the APP gene) which encourages the production of oligomers (that are neurotoxic). These oligomers cluster and eventually form plaques that contribute to symptoms presented above for AD [34,35]. Tau, usually functioning to stabilise axonal microtubules, is abnormally phosphorylated in AD. When this occurs, these highly phosphorylated tau proteins tend to clump together into filaments and further into insoluble neurofibrillary aggregates, spreading throughout the brain [36,37]. Tauopathy is also closely linked to granulo-vacuolar degeneration (GVD) in AD patients. Granulo-vacuolar bodies (GVBs) are often present in hippocampal pyramidal cells of those with AD, which is likely associated with cognitive decline in patients [34,38,39].
The increased loss of neurons in the substania nigra pars compacta (mostly), combined with the presence of abnormal aggregates, underpins the pathophysiology of PD [40]. Losing these neurons, predominately dopaminergic neurons, reduces dopamine levels, thus leading to the symptoms [41]. Drilling down into mechanisms, alterations in α-synuclein lead to either an increase in aggregation of protein or diminished capacity for its degradation, creating fibrilization of α-synuclein in Lewy bodies or neurites promoting neurodegeneration [42,43]. Furthermore, this α-synuclein accumulation directly increases levels of mitochondrial stress and reactive oxygen species [44,45]. Parkin is associated with many subcellular compartments such as synaptic vesicles and the endoplasmic reticulum, as well as playing a neuroprotective role via mitochondria by delaying mitochondrial swelling and the activation of caspase 3 [46]. Its function is closely linked to the ubiquitin-proteasome proteolytic pathway. Parkin is an E3 ubiquitin-protein ligase that, in partnership with E2-conjugating enzymes, selects protein substrates for ubiquitylation and their subsequent degradation [46,47,48]. Oxidative stress transforms DJ-1 (which has a key role in antioxidant activities as well as in directly inhibiting α-synuclein aggregation) into a more acidic isoform which translocates to the outer mitochondrial membrane with PINK1 (located on the inner membrane of mitochondria), regulating the function of mitochondria through phosphorylation of substrates. In neurodegeneration associated with PD, one or more of these pathways is impaired; losing parkin results in the accumulation of mitochondrial substrates, exposing mitochondria to increased stress, and proteasomal impairment amplifies this effect. A reduction in ATP exacerbates the proteasomal impairment, which leads to an increase in α-synuclein accumulation [43,49,50,51,52,53] and a further increase in neurodegeneration.

3. Current Therapeutic Approaches

Treatment of both AD and PD has evolved over time; however, it is still true that no effective treatment for neurodegeneration exists despite extensive time, money and effort being applied to treatment discovery. Researchers have developed a number of models for investigating both diseases, and it is important to understand current treatments, how they work and ways that they can be improved.

3.1. Current Therapeutic Approaches—Alzheimer’s Disease

3.1.1. Pharmacology-Based Therapies

Treatment options targeting AD-associated cognitive impairment have expanded over the last decades with several FDA-approved pharmacological agents being developed. The current FDA-approved treatments for AD are limited to cholinesterase inhibitors (AChEIs) and an N-methyl-D-aspartate (NMDA) receptor antagonist [54].
Targeting acetylcholinesterase (AChE) was established to be the most consistent and viable therapeutic avenue in AD with cholinergic deficits [55]. AChEIs act by blocking cholinesterase enzymes (AChE and butyrylcholinesterase (BChE)) prolonging the half-life of ACh, leading to increased ACh levels available at the synaptic cleft [56]. There are currently three AChE-focused treatments available including galantamine, rivastigmine and donepezil, associated with different time points along the life cycle of AD [55,57]. However, despite initial success with these drugs, generating new, more effective AChEIs failed in clinical trials and has been abandoned due to low efficacy or significant side effects [55]. In more recent years, research has turned to both natural AChEIs (as opposed to synthetic) and hybrids (combining multiple AChEIs together), as a mechanism for overcoming some of the previous shortcomings [58].
Increased N-methyl-D-aspartate receptors (NMDAR) in AD patients results in altered calcium signalling in the brain, causing cell death and dysfunction. Receptor antagonists designed specifically to interact with NMDARs act by reducing calcium alterations and restoration of normal physiological function, ultimately slowing the progression of AD [59,60]. As with AChEIs, these agents are also an approved treatment for PD [59].

3.1.2. Surgery-Based Therapies

There have been several surgical approaches trialled for the treatment of AD, with minimal success and mostly focused on symptom management [61]. Cerebrospinal spinal fluid shunting was thought to increase cerebral blood flow, thus increasing the nutrient supply to affected areas. By doing this, nutrients could support damaged but partially functioning neurons. However, results reported were often subjective with brief follow-up intervals [62]. More recently, this work has focused on the clearance of tau and beta amyloid; however, one study showed no effect on clearance of either protein [63]. Intraventricular infusions have also been trialled as a surgical intervention, but with limited publication of results. Most often the drugs trialled were cholinergic agents such as nerve growth factors or neuroprotective factors such as monosialitetrhexosylgangioside [61]. Lastly, tissue grafts have also been investigated as potential therapeutics in AD; however, there have been few human trials published in this area and more rigorous studies would be needed [64,65].

3.2. Current Therapeutic Approaches—Parkinson’s Disease

3.2.1. Pharmacology-Based Therapies

There have been numerous therapeutic pathways highlighted as targets in PD, which are largely designed to tackle the specific symptoms [66,67]. AChE inhibitors are also used in the treatment of PD, focusing on PD-related dementia, with extremely variable outcomes [68]. One study noted that after the addition of AChEIs there was an improvement in gait parameters, such as swing duration and gait cycle duration in people with Parkinson’s disease (PwP) [69]. A systematic review of the use of AChEIs in PD was conducted in 2014 that theorised that AChEIs could be a positive treatment option for cognitive impairment, but it would need to be balanced with some of the adverse drug reactions observed [70].
Current pharmacological approaches include levodopa, L-DOPA, L-3,4 dihydroxyphenylalanine, dopamine agonists, amantadine, apomorphine or MAO-B inhibitors like selegiline, all of which aim to short-circuit the lack of dopamine in the substantia nigra (SN) [71]. Pharmacological interventions have a complicated relationship with the disease, often producing initial positive results, but most agents only address the dopaminergic aspects of PD and leave its progressive course unaltered [71,72]. Studies have shown that more regular and targeted dosing is required after 3–5 years of the disease and the progressive loss of independence, dyskinesia, confusion and hallucinations [73,74]. There are also reported side effects associated with pharmacological interventions including oxidative stress in patients, gastrointestinal problems, nausea and peripheral oedema [71,72,75,76,77].
Memantine is a partial NMDAR antagonist and has been suggested as a potential new treatment for both AD and PD. Blockage of the NMDA glutamate receptors can normalize the glutamatergic system balance, resulting in improvements in memory and cognitive function, as demonstrated in various animal models and some small patient studies [59,78]. However, its effects appear to have more benefits overall for PD pathologies, with only modest side effects reported such as nausea and drowsiness [59].

3.2.2. Surgery-Based Therapies

Deep brain stimulation offers another avenue of treatment for the improvement of both motor and non-motor symptoms in severe cases of AD and PD with a focus on reducing Lewy bodies, neurofibrillary tangles and β-amyloid 42 oligomers [79,80], yet it is still unclear which targeted brain regions yield the most beneficial effects or which will impact quality of life most [81]. In the case of AD, deep brain stimulation does not appear to be of benefit to early onset AD but did show some promising results in later stages of AD [82].

3.2.3. Physical Therapies

Physical therapy approaches support patients in adjusting their life after diagnosis, including speech therapy, lifestyle changes and cognitive behavioural therapy for both AD and PD [83,84]. Physical therapy helps to improve mood, decrease aggression and improve mobility and strength [85,86].
On the horizon is a more novel therapeutic intervention, with stem cells presenting an exciting opportunity to not only repair damage associated with the loss of dopaminergic neurons but to also adjust the dopamine pathways, reduce tauopathy and tackle neurofibrillary tangles [87,88,89].

4. Potential Future Novel Therapeutic Interventions

While there have been additions and advances to the therapeutic arsenal for AD and PD, there are still many deficits in creating a cohesive and comprehensive treatment plan. Most treatments focus on a specific aspect of the pathology and fall victim to the inability to cross the blood–brain barrier (BBB); however, as we uncover more information about the impact of the BBB in AD and PD, researchers will be assisted in increasing the potency of drugs by utilising targeted delivery systems [90]. There are still many questions unanswered despite all the current knowledge available on the short- to medium-term efficacy of current therapeutic approaches. There is a lack of treatments that diminish the common side effects of nausea, swelling and vomiting, as well as for a targeted treatment that can focus on a specific pillar pathway in the disease architecture. Lastly, there is a need for treatments that not only restore the losses observed in AD and PD but can also prevent further damage.
Recent research has uncovered a potential positive therapeutic effect of nanoparticles for disease therapies, with many small particles including metals, C NPs and exosomes being used in the treatment of neurodegeneration [91]. Extracellular vesicles (EVs), cell-penetrating peptides (CPPs) and microRNAs (miRNAs) have been shown to have immense potential in neurodegeneration. EVs are seen as carriers for small amounts of genetic information or other small molecules delivered via local or systemic routes [92]. Over the past decade, a tremendous amount of research has highlighted the potential of EVs. They have both the potential to ameliorate disease and may be suitable as the next generation therapeutic vehicle [93]. In parallel, cell-penetrating peptides have been discovered which are short, positively charged, amino acid sequences which can penetrate cell membranes [94]. They can translocate small macromolecules such as nucleic acids across the cell membrane as CPP+ cargo complexes [95]. miRNAs have been reported to function as mediators of cell–cell communication; however, their main function is post-transcriptional gene expression regulation [96]. Furthermore, there is more research to suggest that by using CPP+ cargo complexes, miRNAs can activate genes under certain conditions [97]. There is an exciting new era in prospect by utilising these approaches, either individually or combined, and their potential novel therapeutic effect in the field of neurodegeneration. In this review, we explore the potential use and combination of extracellular vesicles, cell-penetrating peptides and microRNAs (miRNAs) as novel therapeutic interventions for treatment of PD and AD.

4.1. Extracellular Vesicles in Neurodegeneration

EVs are membranous nanoparticles secreted by almost all types of cells (both animals and plants) into the extracellular space [98,99,100,101]. EVs have been associated with several roles within the central nervous system, including positive effects on cell function, survival and intracellular communication and negative effects through the detrimental spread of proteins associated with neurodegeneration [100,101].
There are three major classes of EVs: exosomes, ectosomes and apoptotic bodies. Exosomes are 40–160 nm in diameter and are typically derived from both the endocytic pathway and the Golgi apparatus. Ectosomes are more diverse at 50 nm–1 µm in diameter and derived from direct liberation of plasma membrane fragments [102]. Lastly, apoptotic bodies range from 50 to 5000 nm in diameter and are generally produced from cells undergoing programmed cell death [103]. The cell membrane or Golgi apparatus (through endocytosis) fuses with endosomes that undergo inward budding [104,105], giving rise to multivesicular bodies (MVBs) [106] that contain intraluminal vesicles (ILVs). These ILVs contain cargo, for example, lipids, proteins, mRNAs and microRNAs [107], which either are degraded by lysosomes or bind to the plasma membrane for release into the extracellular space [108].
Many neurodegenerative diseases share some common features in the molecular mechanisms that underpin their pathogenesis, including protein misfolding and aggregation. The proteins that are misfolded in neurodegenerative diseases often evade usual clearing mechanisms such as refolding and degradation [109,110]. Initially, some studies suggested that EVs were carriers for misfolded or dysfunctional proteins such as α-synuclein in PD and amyloid β-protein in AD. However, this theory has been disputed over the past decade, with a broader view and the potential neuroprotective properties of these vesicles now actively being explored across many neurodegenerative diseases (Figure 2) [111,112].
Extracellular vesicles are known to have a range of positive biological functions including tissue repair, regeneration and immune surveillance [100]. Much interest in EVs has focused on their ability to modulate homeostasis, remove unwanted material and neutralise synaptic plasticity, disrupting for example the negative effects of amyloid β-protein [113]. Moreover, EVs can cross the blood–brain barrier, a shortcoming of many potential therapeutics. This is important in increasing the availability of different molecules of interest that can serve as therapeutics in neurodegeneration. Many studies have investigated the effect of EVs (derived from different sources) on features of the pathogenesis of AD and PD [114]. Mesenchymal stromal cell-derived EVs are the most common source of EVs studied to date, with researchers investigating protein aggregation, cognitive functions and neurodegenerative pathways. Protein aggregation is a major cause of pathology in both AD and PD. Over 20 studies have shown that EVs can reduce neuroinflammation, therefore leading to an associated reduction in the accumulation of Aβ and α-synuclein (SNCA) [115,116]. In addition, three studies have suggested that EVs in AD and PD models have been able to reduce oxidative stress, mitochondrial stress and pro-inflammatory cytokines such as IL-1β and TNF-a, while anti-inflammatory molecules such as YM-1, MRC1, FIZZ1, and CD163 were increased [116,117,118]. EVs have also been described as aiding neurite outgrowth and axon regeneration of damaged neurons in vivo [119,120]. Studies have also shown EVs’ ability to decrease the number/severity of damaged or destroyed neurites, both in vitro and in vivo [121,122]. There is also evidence to suggest that the overall level of apoptosis reduces after the addition of EVs in an in vitro model [123]. Lastly, it has been shown that exosomes derived from bone marrow stem cells reduced impairment associated with AD. After injection of exosomes into STZ-injected mice, researchers found that these exosomes reduced the hyperactivation of microglia and astrocytes as well as reducing IL-1β, IL-6, TNF-α, Aβ1-42, and p-Tau [124].

4.2. Extracellular Vesicles as Cargo Delivery Vehicles

An attractive aspect of EV biology is their ability for cellular uptake as a mechanism for vesicle cargo transport. Several mechanisms of EV uptake have been proposed including direct binding to the surface of the EVs, phagocytosis, micropinocytosis and direct membrane fusion [125]. EVs can be internalized by nearly all cell types in a highly regulated process. Both the target cell and EV must possess the complimentary cell surface receptors and ligands [125,126,127].
There are a number of properties of EVs which suggest they would be effective as cargo vehicles:
  • Their ability to migrate across the BBB, in a bi-directional manner, with several studies showing their proficiency in this area, albeit with very little information about their mechanism of action [128,129,130]. A study evaluating 6-hydroxy-dopamine (6-OHDA)-induced ND on dopaminergic neurons in SH-SH5Y cells found that MSC-derived EVs reduced DA neuronal death in this in vitro model. Furthermore, they found these EVs also reduced toxicity and neuronal death in their rat model [131].
  • They can carry miRNAs. EVs can mediate the movement and delivery of many miRNAs, including miR-21. This miR is usually associated with the microglial anti-inflammatory (although in other cases it has been reported to be inflammatory) response and indicated as a potential new AD biomarker. This has been demonstrated in in vitro studies of SH-SH5Y cells that were transfected with APP [132,133].
  • EVs can be utilized as a cargo vehicle for therapeutic agents that target neurodegeneration. For example, curcumin is a drug that is suggested to reduce neuroinflammation and oxidative stress in AD, yet it lacks the ability to cross the BBB alone. It has been suggested that combining EVs and curcumin may help overcome some of these shortcomings [134,135,136]. In PD, dopamine has been trialled as a potential drug to be carried by EVs to enhance its delivery to cells, which seems to create a less transient expression of dopamine and increased neuronal function in their model [133,137]. Therapeutic proteins are loaded into exosomes ex vivo and can deliver neuroprotective effects on specific cells in the brain, with EVs crossing the BBB. They cross the BBB through targeting endothelial cells via receptor interactions, thus entering the cell through membrane fusion and endocytosis. Catalase loaded into exosomes has shown neuroprotective effects in the microglial cells, with a reduction in microgliosis in their 6OHDA-induced neurodegeneration model, as measured by a reduction in CD111b expression [138]. EVs augmented or transfected with plasmids have also been used in animal models of PD to evaluate their therapeutic effect. EVs with catalase-encoded plasmids have been shown to transverse the BBB, to incorporate with neurons and ameliorate PD symptoms in mouse models [138]. Macrophages that were genetically modified to express glial-derived neurotrophic factor (GDNF) were found to express modified EVs with GDNF and were shown to slow the progression of PD via a reduction in neuro-inflammation [139].
Despite the relative ease of transfer, the size (for example, catalase encapsulated into an EV sized between 100–200 nm showed a reduction in neuroinflammation, but other sized EVs did not show the same catalase uptake rate [138,140]) and the morphology of the EVs are crucial to success and therefore must be carefully selected [138]. It is also important to consider the origin of EVs, as this will dictate some of the neuroprotective effect they may elicit. For instance, macrophage-derived EVs innately target inflamed tissue within PD and AD, suggesting that they may be the best target EV to combine with a therapeutic protein, drug or RNA to tackle neuroinflammation in the brain [141]. There are several components to consider when exploiting EVs as therapeutics in ND.

4.3. Cell-Penetrating Peptides (CPPs) in Neurodegeneration

CPPs are short basic amino acid sequences that facilitate the movement of molecules, mainly proteins into cells and across the blood–brain barrier. They are typically 4–50 residues and are categorised by their physicochemical properties as cationic, amphipathic, or hydrophobic [142,143,144].
CPPs are classified based on the origin of the peptide (synthetic, chimeric or protein-derived) and their physicochemical properties (cationic, hydrophobic or amphipathic) [95]. Most CCPs are classified as cationic due to the presence of a positive charge [95]. There are three possible factors which influence CPP internalisation across membranes: the peptide concentration, the peptide sequence and the lipid components of each membrane [145,146]. Direct membrane penetration occurs at higher peptide concentrations, whereas at lower concentrations internalization occurs through endocytosis [145]. Arginine-rich CPPs yield higher concentrations in bio membranes because of their higher positive charge ratio [95]. Proteoglycans provide the first point of contact between CPPs and the cell surface via electrostatic interaction [145]. This contact initiates selective activation of some GTPases [147] influencing cell membrane fluidity and allowing CPP cell entry [145,148].
To date, a limited number of studies have been performed on the neuroprotective and other beneficial effects of CPPs in AD and PD (Figure 3). For example, gastric inhibitory polypeptide (GIP), a hormone and growth factor playing a role in the brain, has exhibited neuroprotective effects in AD and PD animal models [149]. In addition to the neuroprotective effects of GIP and other similar peptides, including glucagon-like peptide 1 (GLP), the synaptic plasticity was retained and cell repair and mitochondrial functions were restored with the preservation of dopaminergic neuronal function [150,151]. A newer approach with CPPs is engineering CPP antagonists to combat proteins of interest, including amyloid-β [152]. Researchers are additionally exploiting the relative biocompatibility and low cost of CPPs with specific therapeutic effects and efficient delivery [153]. To date studies have had success in using CPPs to inhibit AB oligomerization and neurotoxicity, thus reducing mitochondrial stress, oxidative stress and proteasomal inhibition [142].

4.4. Cell-Penetrating Peptides (CPPs) as Cargo Delivery Vehicles

Intracellular delivery of large molecules suffers from a reduced capacity to cross the BBB and then retention in the brain. However, CPPs have been developed to be conjugated with small molecules such as drugs due to their high affinity for intracellular delivery [154].
CPPs have been shown to efficiently enter cells with a low level of cytotoxicity [155,156], supporting their use for intracellular delivery of cargo [157]. This can help to alleviate not only the limitations involved in conventional drug delivery [157] but also facilitate the delivery of siRNAs and other molecules to the target site [156].
CPPs can facilitate intracellular delivery of proteins through covalent bonds with their cargos. CPPs conjugated with penetrating and TAT show elevated abilities to block caspase activation, important in reducing apoptosis and protecting neurons [157,158]. Tagging or encapsulating enzymes with CPPs aids their movement across the BBB for tackling effects of neurodegeneration including oxidative stress and autophagy. TAT, along with glyoxalase, catalase and CPP with L-asparaginase can prevent oxidative stress and damage to neuronal cells [159]. However, there are concerns over cationic ions that may bind with negatively charged molecules, leading to aggregation, the very thing researchers are trying to avoid in ND, leading to a pivot towards using new approaches like microRNA [160].

4.5. Micro RNAs (miRNAs) in Neurodegeneration

miRNAs are small non-coding RNAs discovered in the early 1990s. They are transcribed from DNA sequences in primary miRNAs, which are then further processed into pre-cursor miRNAs and mature miRNAs [96,161,162]. To date, miRNAs have been uncovered in all animal models, with recent discovery of their roles in gene expression regulation [163,164,165,166,167]. Mostly, miRNAs suppress gene expression by interacting with the 3′ UTR region of a target mRNA, but some will interact with the 5′ UTR, coding sequence or gene promoter region of other mRNAs [163,168]. In some cases, and under certain conditions (for example, with miR369-3p, TNF alpha is recruited into specific microRNAs to promote activation of specific G0 expressed mRNAs), miRNAs can induce gene expression [96,169].
miRNAs have many important roles to play in a range of biological processes. Specific neuronal miRNAs can control neuronal differentiation, excitability and function [170]. miRNA biogenesis is a tightly controlled process regulated at multiple levels. This includes transcription, RNA editing and methylation, adenylation and RNA decay [163]. The 3′ untranslated region (UTR) of mRNAs generally contains miRNA-binding sites, while the 5′ UTR is crucial for target recognition [163]. More than 60% of human protein-coding genes have at least one miRNA binding site, indicating that they may be controlled, to a certain extent, by miRNAs [171]. While this may give us an insight into the pathogenesis of neurodegenerative diseases, it also gives us a therapeutic avenue to explore. miRNAs provide a pathway to study the commonalities in the pathophysiology of both AD and PD. Dysregulated miRNAs in both diseases are primarily associated with apoptosis and inflammation, but miRNAs targeting the regulation of APP, L1CAM and the caspase family could see a reduction in neurodegeneration, as they are some common targets for dysregulation in both AD and PD [172].
Detecting both AD and PD in early stages is problematic, but miRNAs present an alternative approach to early detection, and one which is both cost-effective and non-invasive [173]. Both cerebrospinal fluid (CSF) and blood have been suggested as fluids for biomarker testing, with various studies evaluating these in the context of early detection of AD and PD [173,174]. Some examples include miR-23a and 29b, found in serum and CSF, respectively, being used to detect mild cognition impairment AD [175,176], and changes in circulating miRNAs such as miR-30a-5p and miR-185-5p being used for PD [177,178].
Many miRNAs have been indicated as positive modulators in ameliorating pathogenesis in AD and PD; for example, overexpressing miR-34a has been indicated to reduce tau synthesis in AD by binding to the 3′ UTR of human tau mRNA [179]. There is in vitro and in vivo evidence that miR34b/c regulates DJ-1 and parkin, two key PD genes, as well as for the role that miR-29 plays in neuronal protection [180,181]. With all of this in mind, either replacement miR treatment (to augment the downregulation of key miRNAs in AD and PD), or addition of neuroprotective miRNAs, may unlock new avenues for tackling neurodegeneration. Recently, miRNA 124-3p-enhanced extracellular vesicles have been shown, in vivo, to protect neurons in the substantia nigra, and they have the potential to reduce key pathophysiological problems associated with neurodegeneration such as oxidative stress, mitochondrial stress and superoxide production [182]. Targeting α-synuclein is a major therapeutic target for PD, but also for other cases of neurodegeneration. Many miRNAs have been investigated for their role in the homeostasis of α-synuclein, with several miRNAs being identified to date, including miR-7, miR-34b and miR-214, that decrease α-synuclein levels in vitro and in vivo [183]. The future of miRNAs is bright, and the possibility of exploiting their neuroprotective effects with the cargo abilities of EVs or CPPs offers a new avenue for the treatment of PD and AD.

5. Next Generation of Novel Therapeutics in AD and PD

Significant progress over the last decade has been made in elucidating the potential functions of EVs and CPPs mentioned above, presenting new avenues for diagnostics and therapeutics [95,184]. The research suggests that we can look at EVs and CPPs in two ways. First, they can function as intracellular carriers for therapeutics due to their ability to cross the blood–brain barrier [157,185,186,187]. Second, they could function as the next generation of therapeutics on their own [95,149,188,189].
However, to look at EVs and their intracellular carrier ability, we also have to look at their ability to carry disease [190]. It has been theorised that EVs can potentiate and propagate neurodegenerative pathology via intracellular communication and transport, as well as play a role in the misfolding of key proteins associated with neurodegenerative diseases [101,191,192]. Despite this, exploiting this very mechanism in a combinatory approach with both CPPs and miRNAs could unlock novel neuroprotective therapies (Figure 4).
Conventional methods of drug delivery often incorporate liposomes or synthetic carriers; however, extracellular vesicles have many advantages over these classical methods [187]. EVs are comprised of a mixture of lipids and membrane proteins [193], facilitating the targeting of specific tissues, as well as minimising non-specific interactions [187,193,194]. There is extensive research that supports the cross-talk ability of EVs (with other cells), and the impact this may have on their therapeutic properties. Neurons and microglia communicate bidirectionally, sensing and responding to stimuli around them, and this communication can be mediated by soluble factors such as EVs [195]. The inherent ability of EVs to target specific cells allows the delivery of functional RNA and soluble cytokines involved in the immune response across the blood-brain barrier [196,197]. EVs play roles in many neuronal homeostatic processes such as neuron excitability, synaptic plasticity and microglial activation, often due to their unique intracellular communication pathways and the extensive list of potential cargo molecules [198]. We theorise that modifying the CPPs on the surface of an EV to become more arginine rich (increasing their cell-penetrating capacity), in combination with miRNAs targeting ND (such as miR-7 that targets alpha-synuclein regulation), will increase the neuroprotective effects that EVs, CPPs and miRNAs can have individually, as shown in Figure 4.
This opens an exciting domain in neurodegenerative therapeutics with the possibility of combing EVs with CPPs and miRNAs, exploiting the natural cargo delivery ability of EVs. miRNAs are small non-coding molecules that can regulate gene expression by directing mRNA cleavage or translational inhibition [199,200]. Up to one third of human genes may be regulated by miRNAs, playing roles in the immunological response to several disease pathologies [199]. Therapies involving miRNAs fall into two categories: miRNA inhibition of disease-specific miRNAs and miRNA replacement to encourage disease-free (repressed) miRNA expression [201]. In both PD and AD, dysregulation of non-coding RNAs has been reported [202,203,204]. For example, SNCA dysregulation is a major pathological problem in PD, with several miRNAs being investigated and suggested as potential α-SYN regulators, with studies demonstrating that some miRNAs could protect different cell types from neurodegeneration arising from synucleinopathy [205,206]. These include miR-7 and miR-153 protecting SH-SY5Y cells, differentiated human progenitor ReNcells, ventral midbrain (VM) cells and primary mouse neuron from degeneration from known PD-induced cell lines with MPP+ [207,208]. In AD, miRNAs have also been shown to provide a much more comprehensive approach to diagnosis. miR-222, miR-29c-3p and miR-19b-3p are much clearer biomarkers that can be detected in the biofluid of AD patients [209,210,211,212]. Furthermore, the use of miRNAs as a treatment option for AD is a continually evolving field with microarray analysis in Tg2576 transgenic mice identifying both miR-200b and miR200c as down regulators of Aβ secretion by modulation of mTOR [213].
Nevertheless, there are some limitations to using miRNAs, notably in the delivery of miRNAs to target tissues with specificity. There is scope here to combine both EVs, which possess inherent cell-targeting abilities, with therapeutic miRNAs to provide restorative therapeutic options for PD and AD [214]. In addition, while there are still gaps in our understanding of the mechanism of action of EVs, it is agreed that the delivery of EV cargo creates benefits in the treatment of disease. Taking this into account, it could be theorised to combine EVs and miRNAs together to create a complementary therapeutic, one that utilises EVs’ innate ability to target cells and tissues with specificity along with miRNAs’ neuroprotective effects [214].
Similar to the case with EVs, CPPs also facilitate the intracellular delivery of a variety of bioactive cargos; however, they lack cell specificity and often have a short/limited time of action. Hence, there is a need to optimise the CPP that is optimal for a particular target of interest. Combining CPPs with specific and more capable delivery systems, such as EVs, will improve their clinical enhancements [95,157]. There are some concerns about cytotoxicity related to both CPPs and the “cargo” they may carry—however, data indicates that cytotoxicity is low in their effective dose range, and heavily depends on CPP type and composition. This highlights the importance of selecting the appropriate CPP and concentration [154,155,215,216]. CPPs are limited by low cell and tissue selectivity [217]; however, like miRNAs, EVs may be a solution to this problem.
Recent studies have investigated modifying arginine-rich CPPs on the membranes of EVs as a method for improving effective micropinocytosis and increasing the uptake of cellular EVs expressing CPPs [218,219]. Dodecaborate-encapsulated EVs with modified hexadeca oligoarginine, a CPP, showed an amplification in the actin-dependent endocytotic pathway, micropinocytosis and cellular uptake of the specific EV [220]. This work also highlighted the importance of activating arginine peptide sequences for the efficient release of EV cellular cargo and improving the penetrating ability of these EVs [218]. This is important in the context of combining a triad approach to neurodegeneration where a potential EV could be modified with arginine-based peptides for the more efficient release of miRNAs that target alpha-synuclein in PD and tau in AD.

6. Current Barriers to Clinical Translation

There are still many challenges for the use of EVs and CPPs as cargo vehicles. This is a science in its infancy, and for EVs we lack an understanding of their mechanism of transport across the BBB, their biodistribution, their contents and their pharmacokinetic properties [133]. EVs have a unique surface make-up including protein receptors, transcription factors, enzymes, extracellular matrix proteins, lipids and nucleic acids (DNAs, mRNAs and miRNAs) that are within and on their surface [221]. The easiest method of loading therapeutics into EVs is to mix EVs with free drugs; however, it is difficult to control this loading efficiency and the drug may suffer from degradation within the host cells [222,223]. It is also thought the EVs contain some of their parent material from formation, thus reducing the space available for encapsulation of different drugs/proteins/RNA [140,224,225]. Many methods have been suggested for the incorporation of therapeutics with EVs, including electroporation, sonication, extrusion, and freeze/thaw cycles; however, these come with their own challenges of disrupting the integrity of the exosomes, deformation of EV membranes, exosome aggregation and low yield (with variability between EV batches) [109,226,227,228,229,230].
Incorporating therapeutics such as drugs, proteins and nucleic acids also depends on the cellular processes that produce EVs; it is a delicate balance of ensuring the right cargo molecule for the right EV, often producing variable results in terms of efficacy and potency in its effect [231]. We must also consider the source of EVs; even though nearly all cells produce some type of EV, some are not suitable to be drug carriers. In this instance, the surface proteins, size yield and intracavitary composition are key characteristics to evaluate when selecting the “ideal” EV for cargo delivery [232].
EVs contain proteins both within and on their surface, and many of these have been exploited in therapeutics, including an EV-based antioxidant and catalase delivery system for PD [138]; however, packaging active proteins or RNAs still remains challenging [232]. To date, there is no standardised method of EV extraction (although there are many guidelines from the International Society of Extracellular Vesicles (ISEV)), creating a bottleneck for the use of EVs as drug carriers, as none of the current approaches meets the ideal criteria for isolation, with many limiting factors including low specificity and low recovery in some cases [232,233,234,235].
Our current understanding of the pathophysiology of AD and PD, which is often neuron-centric, is a barrier to the generation of novel therapeutics. For example, targeting amyloid-beta and hyperphosphorylated tau as a therapeutic in AD shows very little clinical relevance despite extensive studies. Recently, work has shown that alterations in the gut microbiome may generate negative consequences for mitochondrial function in the body [236]. This discovery has proved an exciting avenue of research as a treatment target for AD and PD. Is amyloid-beta just “too much of a good thing”, with AD and PD driven by the suppression and/or dysregulation of mitochondria-linked glial processes that would normally dampen local inflammatory processes [237]?
Lastly, there is a lack of clinical translation for the use of EVs as cargo vehicles, and while some work has been done in this area, questions remain about the safety, potency and purity of any clinical products that may be developed [238].
There is less known about CPPs as therapeutics in comparison with EVs, but they are still a potentially exciting new approach to treating neurodegeneration, as they have the capacity to cross many barriers such as the BBB, skin and cornea. However, they also have many limitations, including transport efficiency, target specificity and CPP-cargo coupling [239]. CPPs are likely to be degraded after exposure to biological fluids such as blood, due to their peptidic nature, making the development of such therapeutics difficult [240]. Tracking CPPs is also tricky; it is possible to conjugate them with a fluorophore (GFP, RFP or others), but ultimately this will change the physiochemical properties of the CPP of choice and thus its biodistribution. Detecting non-labelled CPPs is hampered due to a lack of specific antibody availability for techniques such as an enzyme-linked immunosorbent assay [241], while there are also challenges around detecting fluorescently labelled CPPs in vitro, because the fluorescence may be blocked by tryptophan residues of close transmembrane proteins on the membrane bilayer [242].
CPPs are significantly limited by their chemical instability; they are subject to degradation via intra- and extracellular enzymes, subsequently lowering their concentration and thus their potency (although some biodegradability is desirable as it improves biocompatibility and patient safety) [243]. There are many strategies aiming to overcome this excessive degradation, including modifying the amino acid residues at specific sites to reduce access to proteases [244], which seems to have promising results. In order to translate CPPs to clinical research, however, there is a need to have a more robust targeted drug delivery system to ensure efficient delivery and drop off of the CPP and its cargo. In addition to this, there is a need to create robust CPPs that reach their specific target, minimising off-target delivery which could lead to adverse side effects [243].

7. Conclusions

PD and AD are progressive neurodegenerative disorders which represent a substantial unmet clinical need driving the requirement for new therapeutic approaches, focusing on both restorative and protective neuronal therapies. Extracellular vesicles have been tipped to be a leading therapy in this field, though they have some drawbacks in application as a sole therapeutic agent. These include cell target specificity, transport across the blood–brain barrier, and the ability to target multiple pathways of degeneration including oxidative stress, neuronal cell death and mitochondrial dysfunction. These limitations can potentially be overcome by combining the neuroprotective features of EVs, CPPs and miRNAs, which have complimentary features that lend themselves to being fit together in a novel combinational therapeutic agent able to tackle the multiple pathways of neurodegeneration exhibited by both AD and PD.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dugger, B.N.; Dickson, D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kurosinski, P.; Guggisberg, M.; Götz, J. Alzheimer’s and Parkinson’s disease—Overlapping or synergistic pathologies? Trends Mol. Med. 2002, 8, 3–5. [Google Scholar] [CrossRef]
  3. Uttara, B.; Singh, A.V.; Zamboni, P.; Mahajan, R.T. Oxidative Stress and Neurodegenerative Diseases: A Review of Upstream and Downstream Antioxidant Therapeutic Options. Curr. Neuropharmacol. 2009, 7, 65–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tan, S.H.; Karri, V.; Tay, N.W.R.; Chang, K.H.; Ah, H.Y.; Ng, P.Q.; Ho, H.S.; Keh, H.W.; Candasamy, M. Emerging pathways to neurodegeneration: Dissecting the critical molecular mechanisms in Alzheimer’s disease, Parkinson’s disease. Biomed. Pharmacother. 2019, 111, 765–777. [Google Scholar] [CrossRef]
  5. Xie, A.; Gao, J.; Xu, L.; Meng, D. Shared mechanisms of neurodegeneration in Alzheimer’s disease and Parkinson’s disease. Biomed. Res. Int. 2014, 2014, 648740. [Google Scholar] [CrossRef] [PubMed]
  6. Guadagnolo, D.; Piane, M.; Torrisi, M.R.; Pizzuti, A.; Petrucci, S. Genotype-Phenotype Correlations in Monogenic Parkinson Disease: A Review on Clinical and Molecular Findings. Front. Neurol. 2021, 12, 648588. [Google Scholar] [CrossRef]
  7. Kiriyama, Y.; Nochi, H. The Function of Autophagy in Neurodegenerative Diseases. Int. J. Mol. Sci. 2015, 16, 26797–26812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Scorziello, A.; Borzacchiello, D.; Sisalli, M.J.; Di Martino, R.; Morelli, M.; Feliciello, A. Mitochondrial Homeostasis and Signaling in Parkinson’s Disease. Front. Aging Neurosci. 2020, 12, 100. [Google Scholar] [CrossRef] [Green Version]
  9. Dias, V.; Junn, E.; Mouradian, M.M. The Role of Oxidative Stress in Parkinson’s Disease. J. Parkinsons Dis. 2013, 3, 461–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Gan, L.; Cookson, M.R.; Petrucelli, L.; La Spada, A.R. Converging pathways in neurodegeneration, from genetics to mechanisms. Nat. Neurosci. 2018, 21, 1300–1309. [Google Scholar] [CrossRef]
  11. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef] [PubMed]
  12. Apostolova, L.G. Alzheimer Disease. Continuum 2016, 22, 419–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wattmo, C.; Minthon, L.; Wallin, K. Mild versus moderate stages of Alzheimer’s disease: Three-year outcomes in a routine clinical setting of cholinesterase inhibitor therapy. Alzheimer Res. Ther. 2016, 8, 7. [Google Scholar] [CrossRef] [Green Version]
  14. Dubois, B.; Hampel, H.; Feldman, H.H.; Scheltens, P.; Aisen, P.; Andrieu, S.; Bakardjian, H.; Benali, H.; Bertram, L.; Blennow, K.; et al. Preclinical Alzheimer’s disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016, 12, 292–323. [Google Scholar] [CrossRef]
  15. McKhann, G.; Drachman, D.; Folstein, M.; Katzman, R.; Price, D.; Stadlan, E.M. Clinical diagnosis of Alzheimer’s disease: Report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 1984, 34, 939–944. [Google Scholar] [CrossRef] [Green Version]
  16. Neugroschl, J.; Wang, S. Alzheimer’s disease: Diagnosis and treatment across the spectrum of disease severity. Mt. Sinai J. Med. 2011, 78, 596–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
  18. Singh, S.K.; Srivastav, S.; Yadav, A.K.; Srikrishna, S.; Perry, G. Overview of alzheimer’s disease and some therapeutic approaches targeting aβ by using several synthetic and herbal compounds. Oxid. Med. Cell. Longev. 2016, 2016, 7361613. [Google Scholar] [CrossRef] [Green Version]
  19. Spires-Jones, T.L.; Hyman, B.T. The Intersection of Amyloid Beta and Tau at Synapses in Alzheimer’s Disease. Neuron 2014, 82, 756–771. [Google Scholar] [CrossRef] [Green Version]
  20. Armstrong, R.A. Risk factors for Alzheimer’s disease. Folia Neuropathol. 2019, 57, 87–105. [Google Scholar] [CrossRef] [Green Version]
  21. Emamzadeh, F.N.; Surguchov, A. Parkinson’s disease: Biomarkers, treatment, and risk factors. Front. Neurosci. 2018, 12, 612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Armstrong, M.J.; Okun, M.S. Diagnosis and treatment of parkinson disease: A review. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef] [PubMed]
  23. Braak, H.; Del Tredici, K.; Rüb, U.; de Vos, R.A.I.; Jansen Steur, E.N.H.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef] [PubMed]
  24. Serrano-Pozo, A.; Frosch, M.P.; Masliah, E.; Hyman, B.T. Neuropathological Alterations in Alzheimer Disease. Cold Spring Harb. Perspect. Med. 2011, 1, a006189. [Google Scholar] [CrossRef] [Green Version]
  25. Stoker, T.B.; Torsney, K.M.; Barker, R.A. Emerging Treatment Approaches for Parkinson’s Disease. Front. Neurosci. 2018, 12, 693. [Google Scholar] [CrossRef] [Green Version]
  26. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
  27. El-Agnaf, O.M.A.; Salem, S.A.; Paleologou, K.E.; Curran, M.D.; Gibson, M.J.; Court, J.A.; Schlossmacher, M.G.; Allsop, D. Detection of oligomeric forms of α-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006, 20, 419–425. [Google Scholar] [CrossRef]
  28. Bekris, L.M.; Mata, I.F.; Zabetian, C.P. The genetics of Parkinson disease. J. Geriatr. Psychiatry Neurol. 2010, 23, 228–242. [Google Scholar] [CrossRef]
  29. Simón-Sánchez, J.; Schulte, C.; Bras, J.M.; Sharma, M.; Gibbs, J.R.; Berg, D.; Paisan-Ruiz, C.; Lichtner, P.; Scholz, S.W.; Hernandez, D.G.; et al. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat. Genet. 2009, 41, 1308–1312. [Google Scholar] [CrossRef]
  30. Piaceri, I.; Nacmias, B.; Sorbi, S. Genetics of familial and sporadic Alzheimer s disease. Front. Biosci. 2013, E5, 167–177. [Google Scholar] [CrossRef] [Green Version]
  31. Avazzadeh, S.; Baena, J.M.; Keighron, C.; Feller-Sanchez, Y.; Quinlan, L.R. Modelling Parkinson’s Disease: iPSCs towards Better Understanding of Human Pathology. Brain Sci. 2021, 11, 373. [Google Scholar] [CrossRef] [PubMed]
  32. Morgan, S.L.; Naderi, P.; Koler, K.; Pita-Juarez, Y.; Prokopenko, D.; Vlachos, I.S.; Tanzi, R.E.; Bertram, L.; Hide, W.A. Most pathways can be related to the pathogenesis of alzheimer’s disease. Front. Aging Neurosci. 2022, 14, 846902. [Google Scholar] [CrossRef] [PubMed]
  33. Ahmad, K.; Baig, M.H.; Mushtaq, G.; Kamal, M.A.; Greig, N.H.; Choi, I. Commonalities in Biological Pathways, Genetics, and Cellular Mechanism between Alzheimer Disease and Other Neurodegenerative Diseases: An In Silico-Updated Overview. Curr. Alzheimer Res. 2017, 14, 1190–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185. [Google Scholar] [CrossRef]
  35. De Strooper, B.; Vassar, R.; Golde, T. The secretases: Enzymes with therapeutic potential in Alzheimer disease. Nat. Rev. Neurol. 2010, 6, 99–107. [Google Scholar] [CrossRef] [Green Version]
  36. Braak, H.; Alafuzoff, I.; Arzberger, T.; Kretzschmar, H.; Del Tredici, K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006, 112, 389–404. [Google Scholar] [CrossRef] [Green Version]
  37. Alonso, A.D.C.; Grundke-Iqbal, I.; Iqbal, K. Alzheimer’s disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules. Nat. Med. 1996, 2, 783–787. [Google Scholar] [CrossRef]
  38. Wiersma, V.I.; Hoozemans, J.J.M.; Scheper, W. Untangling the origin and function of granulovacuolar degeneration bodies in neurodegenerative proteinopathies. Acta Neuropathol. Commun. 2020, 8, 153. [Google Scholar] [CrossRef]
  39. Castellani, R.J.; Gupta, Y.; Sheng, B.; Siedlak, S.L.; Harris, P.L.; Coller, J.M.; Perry, G.; Lee, H.-G.; Tabaton, M.; Smith, M.A.; et al. A novel origin for granulovacuolar degeneration in aging and Alzheimer’s disease: Parallels to stress granules. Lab. Investig. 2011, 91, 1777–1786. [Google Scholar] [CrossRef] [Green Version]
  40. Lang, A.E.; Lozano, A.M. Parkinson’s disease. First of two parts. N. Engl. J. Med. 1998, 339, 1044–1053. [Google Scholar] [CrossRef]
  41. Hoang, Q.Q. Pathway for Parkinson disease. Proc. Natl. Acad. Sci. USA 2014, 111, 2402–2403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Shastry, B.S. Parkinson disease: Etiology, pathogenesis and future of gene therapy. Neurosci. Res. 2001, 41, 5–12. [Google Scholar] [CrossRef] [PubMed]
  43. Corti, O.; Hampe, C.; Darios, F.; Ibanez, P.; Ruberg, M.; Brice, A. Parkinson’s disease: From causes to mechanisms. Comptes Rendus Biol. 2005, 328, 131–142. [Google Scholar] [CrossRef] [PubMed]
  44. Park, J.-H.; Burgess, J.D.; Faroqi, A.H.; DeMeo, N.N.; Fiesel, F.C.; Springer, W.; Delenclos, M.; McLean, P.J. Alpha-synuclein-induced mitochondrial dysfunction is mediated via a sirtuin 3-dependent pathway. Mol. Neurodegener. 2020, 15, 5. [Google Scholar] [CrossRef] [Green Version]
  45. Devi, L.; Raghavendran, V.; Prabhu, B.M.; Avadhani, N.G.; Anandatheerthavarada, H.K. Mitochondrial Import and Accumulation of alpha-Synuclein Impair Complex I in Human Dopaminergic Neuronal Cultures and Parkinson Disease Brain. J. Biol. Chem. 2008, 283, 9089–9100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Darios, F.; Corti, O.; Hampe, C.; Muriel, M.-P.; Abbas, N.; Gu, W.-J.; Lücking, C.B.; Hirsch, E.C.; Rooney, T.; Ruberg, M.; et al. Parkin prevents mitochondrial swelling and cytochrome c release in mitochondria-dependent cell death. Hum. Mol. Genet. 2003, 12, 517–526. [Google Scholar] [CrossRef]
  47. Zhang, Y.; Gao, J.; Chung, K.K.K.; Huang, H.; Dawson, V.L.; Dawson, T.M. Parkin functions as an E2-dependent ubiquitin- protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc. Natl. Acad. Sci. USA 2000, 97, 13354–13359. [Google Scholar] [CrossRef] [Green Version]
  48. Grimaldo, L.; Sandoval, A.; Garza-López, E.; Felix, R. Involvement of Parkin in the ubiquitin proteasome system-mediated degradation of N-type voltage-gated Ca2+ channels. PLoS ONE 2017, 12, e0185289. [Google Scholar] [CrossRef] [Green Version]
  49. Healy, D.G.; Abou-Sleiman, P.M.; Valente, E.M.; Gilks, W.P.; Bhatia, K.; Quinn, N.; Lees, A.J.; Wood, N.W. DJ-1 mutations in Parkinson’s disease. J. Med. Genet. 2004, 41, 248. [Google Scholar]
  50. Höglinger, G.U.; Carrard, G.; Michel, P.P.; Medja, F.; Lombès, A.; Ruberg, M.; Friguet, B.; Hirsch, E. Dysfunction of mitochondrial complex I and the proteasome: Interactions between two biochemical deficits in a cellular model of Parkinson’s disease. J. Neurochem. 2003, 86, 1297–1307. [Google Scholar] [CrossRef]
  51. McNaught, K.S.; Jenner, P. Proteasomal function is impaired in substantia nigra in Parkinson’s disease. Neurosci. Lett. 2001, 297, 191–194. [Google Scholar] [CrossRef]
  52. Abou-Sleiman, P.M.; Healy, D.G.; Quinn, N.; Lees, A.J.; Wood, N.W. The role of pathogenicDJ-1 mutations in Parkinson’s disease. Ann. Neurol. 2003, 54, 283–286. [Google Scholar] [CrossRef] [PubMed]
  53. Yang, J.; Kim, K.S.; Iyirhiaro, G.O.; Marcogliese, P.C.; Callaghan, S.M.; Qu, D.; Kim, W.J.; Slack, R.S.; Park, D.S. DJ-1 modulates the unfolded protein response and cell death via upregulation of ATF4 following ER stress. Cell Death Dis. 2019, 10, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Szeto, J.Y.; Lewis, S.J. Current Treatment Options for Alzheimer’s Disease and Parkinson’s Disease Dementia. Curr. Neuropharmacol. 2016, 14, 326–338. [Google Scholar] [CrossRef] [PubMed]
  55. Mehta, M.; Adem, A.; Sabbagh, M. New Acetylcholinesterase Inhibitors for Alzheimer’s Disease. Int. J. Alzheimers Dis. 2012, 2012, 728983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Colović, M.B.; Krstić, D.Z.; Lazarević-Pašti, T.D.; Bondžić, A.M.; Vasić, V.M. Acetylcholinesterase inhibitors: Pharmacology and toxicology. Curr. Neuropharmacol. 2013, 11, 315–335. [Google Scholar] [CrossRef] [Green Version]
  57. Grossberg, G.T. Cholinesterase Inhibitors for the Treatment of Alzheimer’s Disease. Current Ther. Res. 2003, 64, 216–235. [Google Scholar]
  58. Sharma, K. Cholinesterase inhibitors as Alzheimer’s therapeutics (Review). Mol. Med. Rep. 2019, 20, 1479–1487. [Google Scholar] [CrossRef] [Green Version]
  59. Olivares, D.; Deshpande, V.K.; Shi, Y.; Lahiri, D.K.; Greig, N.H.; Rogers, J.T.; Huang, X. N-methyl D-aspartate (NMDA) receptor antagonists and memantine treatment for Alzheimer’s disease, vascular dementia and Parkinson’s disease. Curr. Alzheimer Res. 2012, 9, 746–758. [Google Scholar] [CrossRef]
  60. Liu, J.; Chang, L.; Song, Y.; Li, H.; Wu, Y. The Role of NMDA Receptors in Alzheimer’s Disease. Front. Neurosci. 2019, 13, 43. [Google Scholar] [CrossRef] [Green Version]
  61. Laxton, A.W.; Stone, S.; Lozano, A.M. The Neurosurgical Treatment of Alzheimer’s Disease: A Review. Ster. Funct. Neurosurg. 2014, 92, 269–281. [Google Scholar] [CrossRef] [PubMed]
  62. Adams, R.D.; Fisher, C.M.; Hakim, S.; Ojemann, R.G.; Sweet, W.H. Symptomatic occult hydrocephalus with “normal” cerebrospinal-fluid pressure. A treatable syndrome. N. Engl. J. Med. 1965, 273, 117–126. [Google Scholar] [CrossRef] [PubMed]
  63. Hulstaert, F.; Blennow, K.; Ivanoiu, A.; Schoonderwaldt, H.C.; Riemenschneider, M.; De Deyn, P.P.; Bancher, C.; Cras, P.; Wiltfang, J.; Mehta, P.D.; et al. Improved discrimination of AD patients using beta-amyloid(1-42) and tau levels in CSF. Neurology 1999, 52, 1555–1562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Goldsmith, H.S.; Wu, W.; Zhong, J.; Edgar, M. Omental transposition to the brain as a surgical method for treating Alzheimer’s disease. Neurol. Res. 2003, 25, 625–634. [Google Scholar] [CrossRef] [PubMed]
  65. Goldsmith, H.S. A new approach to the treatment of Alzheimer’s disease: The need for a controlled study. J. Alzheimers Dis. 2011, 25, 209–212. [Google Scholar] [CrossRef] [Green Version]
  66. DeMaagd, G.; Philip, A. Parkinson’s disease and its management: Part 1: Disease entity, risk factors, pathophysiology, clinical presentation, and diagnosis. P T 2015, 40, 504–532. [Google Scholar] [PubMed]
  67. Jankovic, J.; Aguilar, L.G. Current approaches to the treatment of Parkinson’s disease. Neuropsychiatr. Dis. Treat. 2008, 4, 743–757. [Google Scholar] [CrossRef] [Green Version]
  68. van Laar, T.; De Deyn, P.P.; Aarsland, D.; Barone, P.; Galvin, J.E. Effects of cholinesterase inhibitors in Parkinson’s disease dementia: A review of clinical data. CNS Neurosci. Ther. 2011, 17, 428–441. [Google Scholar] [CrossRef]
  69. Tysnes, O.-B.; Storstein, A. Epidemiology of Parkinson’s disease. J. Neural. Transm. 2017, 124, 901–905. [Google Scholar] [CrossRef]
  70. Pagano, G.; Rengo, G.; Pasqualetti, G.; Femminella, G.D.; Monzani, F.; Ferrara, N.; Tagliati, M. Cholinesterase inhibitors for Parkinson’s disease: A systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2014, 86, 767–773. [Google Scholar] [CrossRef]
  71. Zahoor, I.; Shafi, A.; Haq, E. Pharmacological treatment of parkinson’s disease. In Parkinson’s Disease: Pathogenesis and Clinical Aspects; Stoker, T.B., Greenl, J.C., Eds.; Codon Publications: Brisbane, Australia, 2018. [Google Scholar]
  72. Schapira, A.H.V. Present and future drug treatment for Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 2005, 76, 1472–1478. [Google Scholar] [CrossRef]
  73. Carrarini, C.; Russo, M.; Dono, F.; Di Pietro, M.; Rispoli, M.G.; Di Stefano, V.; Ferri, L.; Barbone, F.; Vitale, M.; Thomas, A.; et al. A Stage-Based Approach to Therapy in Parkinson’s Disease. Biomolecules 2019, 9, 388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Antonini, A.; Odin, P.; Pahwa, R.; Aldred, J.; Alobaidi, A.; Jalundhwala, Y.J.; Kukreja, P.; Bergmann, L.; Inguva, S.; Bao, Y.; et al. The Long-Term Impact of Levodopa/Carbidopa Intestinal Gel on ‘Off’-time in Patients with Advanced Parkinson’s Disease: A Systematic Review. Adv. Ther. 2021, 38, 2854–2890. [Google Scholar] [CrossRef]
  75. Hurtig, H.I. Problems with Current Pharmacologic Treatment of Parkinson’s Disease. Exp. Neurol. 1997, 144, 10–16. [Google Scholar] [CrossRef] [PubMed]
  76. Ahlskog, J.E. Treatment of early Parkinson’s disease: Are complicated strategies justified? Mayo Clin. Proc. 1996, 71, 659–670. [Google Scholar] [CrossRef] [PubMed]
  77. Mouchaileh, N.; Hughes, A.J. Pharmacological management of Parkinson’s disease in older people. J. Pharm. Pract. Res. 2020, 50, 445–454. [Google Scholar] [CrossRef]
  78. Companys-Alemany, J.; Turcu, A.L.; Bellver-Sanchis, A.; Loza, M.I.; Brea, J.M.; Canudas, A.M.; Leiva, R.; Vázquez, S.; Pallàs, M.; Griñán-Ferré, C. A Novel NMDA Receptor Antagonist Protects against Cognitive Decline Presented by Senescent Mice. Pharmaceutics 2020, 12, 284. [Google Scholar] [CrossRef] [Green Version]
  79. Bronstein, J.M.; Tagliati, M.; Alterman, R.L.; Lozano, A.M.; Volkmann, J.; Stefani, A.; Horak, F.B.; Okun, M.S.; Foote, K.D.; Krack, P.; et al. Deep brain stimulation for Parkinson disease: An expert consensus and review of key issues. Arch. Neurol. 2011, 68, 165. [Google Scholar] [CrossRef]
  80. Herzog, J.; Fietzek, U.; Hamel, W.; Morsnowski, A.; Steigerwald, F.; Schrader, B.; Weinert, D.M.; Pfister, G.; Müller, D.; Mehdorn, H.M.; et al. Most effective stimulation site in subthalamic deep brain stimulation for Parkinson’s disease. Mov. Disord. 2004, 19, 1050–1054. [Google Scholar] [CrossRef]
  81. Lozano, A.M.; Lipsman, N.; Bergman, H.; Brown, P.; Chabardes, S.; Chang, J.W.; Matthews, K.; McIntyre, C.C.; Schlaepfer, T.E.; Schulder, M.; et al. Deep brain stimulation: Current challenges and future directions. Nat. Rev. Neurol. 2019, 15, 148–160. [Google Scholar] [CrossRef]
  82. Luo, Y.; Sun, Y.; Tian, X.; Zheng, X.; Wang, X.; Li, W.; Wu, X.; Shu, B.; Hou, W. Deep Brain Stimulation for Alzheimer’s Disease: Stimulation Parameters and Potential Mechanisms of Action. Front. Aging Neurosci. 2021, 13, 619543. [Google Scholar] [CrossRef] [PubMed]
  83. Logsdon, R.G.; McCurry, S.M.; Teri, L. Evidence-Based Interventions to Improve Quality of Life for Individuals with Dementia. Alzheimers Care Today 2007, 8, 309–318. [Google Scholar] [PubMed]
  84. Vieregge, P.; Dethlefsen, J. Physical therapy and speech therapy in Parkinson syndrome—A status assessment. Fortschr. Neurol. Psychiatr. 1992, 60, 369–374. [Google Scholar] [CrossRef]
  85. De la Rosa, A.; Olaso-Gonzalez, G.; Arc-Chagnaud, C.; Millan, F.; Salvador-Pascual, A.; García-Lucerga, C.; Blasco-Lafarga, C.; Garcia-Dominguez, E.; Carretero, A.; Correas, A.G.; et al. Physical exercise in the prevention and treatment of Alzheimer’s disease. J. Sport Health Sci. 2020, 9, 394–404. [Google Scholar] [CrossRef] [PubMed]
  86. Borrione, P.; Tranchita, E.; Sansone, P.; Parisi, A. Effects of physical activity in Parkinson’s disease: A new tool for rehabilitation. World J. Methodol. 2014, 4, 133–143. [Google Scholar] [CrossRef] [PubMed]
  87. Duncan, T.; Valenzuela, M. Alzheimer’s disease, dementia, and stem cell therapy. Stem. Cell Res. Ther. 2017, 8, 111. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, Z.; Cheung, H.-H. Stem Cell-Based Therapies for Parkinson Disease. Int. J. Mol. Sci. 2020, 21, 8060. [Google Scholar] [CrossRef]
  89. Liu, X.-Y.; Yang, L.-P.; Zhao, L. Stem cell therapy for Alzheimer’s disease. World J. Stem Cells 2020, 12, 787–802. [Google Scholar] [CrossRef]
  90. Gosselet, F.; Loiola, R.A.; Roig, A.; Rosell, A.; Culot, M. Central nervous system delivery of molecules across the blood-brain barrier. Neurochem. Int. 2021, 144, 104952. [Google Scholar] [CrossRef]
  91. Nguyen, D.D.; Lai, J.-Y. Synthesis, bioactive properties, and biomedical applications of intrinsically therapeutic nanoparticles for disease treatment. Chem. Eng. J. 2022, 435, 134970. [Google Scholar] [CrossRef]
  92. Veziroglu, E.M.; Mias, G.I. Characterizing Extracellular Vesicles and Their Diverse RNA Contents. Front. Genet. 2020, 11, 700. [Google Scholar] [CrossRef] [PubMed]
  93. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
  94. Jain, A.; Jain, S.K. Application potential of engineered liposomes in tumor targeting. In Multifunctional Systems for Combined Delivery, Biosensing and Diagnostics; Elsevier: Amsterdam, The Netherlands, 2017; pp. 171–191. [Google Scholar]
  95. Xie, J.; Bi, Y.; Zhang, H.; Dong, S.; Teng, L.; Lee, R.J.; Yang, Z. Cell-Penetrating Peptides in Diagnosis and Treatment of Human Diseases: From Preclinical Research to Clinical Application. Front. Pharmacol. 2020, 11, 697. [Google Scholar] [CrossRef] [PubMed]
  96. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [Green Version]
  97. Vasudevan, S. Posttranscriptional Upregulation by MicroRNAs. Wiley Interdiscip. Rev. RNA 2012, 3, 311–330. [Google Scholar] [CrossRef]
  98. Robinson, D.G.; Ding, Y.; Jiang, L. Unconventional protein secretion in plants: A critical assessment. Protoplasma 2016, 253, 31–43. [Google Scholar] [CrossRef]
  99. Deatherage, B.L.; Cookson, B.T. Membrane Vesicle Release in Bacteria, Eukaryotes, and Archaea: A Conserved yet Underappreciated Aspect of Microbial Life. Infect. Immun. 2012, 80, 1948–1957. [Google Scholar] [CrossRef] [Green Version]
  100. Croese, T.; Furlan, R. Extracellular vesicles in neurodegenerative diseases. Mol. Asp. Med. 2018, 60, 52–61. [Google Scholar] [CrossRef]
  101. Hill, A.F. Extracellular Vesicles and Neurodegenerative Diseases. J. Neurosci. 2019, 39, 9269–9273. [Google Scholar] [CrossRef]
  102. Doyle, L.; Wang, M. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  103. Kakarla, R.; Hur, J.; Kim, Y.J.; Kim, J.; Chwae, Y.-J. Apoptotic cell-derived exosomes: Messages from dying cells. Exp. Mol. Med. 2020, 52, 1–6. [Google Scholar] [CrossRef] [Green Version]
  104. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. Transport into the Cell from the Plasma Membrane: Endocytosis; Garland Science: New York, NY, USA, 2002. [Google Scholar]
  105. Huotari, J.; Helenius, A. Endosome maturation. EMBO J. 2011, 30, 3481–3500. [Google Scholar] [CrossRef]
  106. Meldolesi, J. Exosomes and Ectosomes in Intercellular Communication. Curr. Biol. 2018, 28, R435–R444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef] [Green Version]
  108. Kalani, A.; Tyagi, A.; Tyagi, N. Exosomes: Mediators of Neurodegeneration, Neuroprotection and Therapeutics. Mol. Neurobiol. 2014, 49, 590–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Li, Z.; Zhou, X.; Wei, M.; Gao, X.; Zhao, L.; Shi, R.; Sun, W.; Duan, Y.; Yang, G.; Yuan, L. In Vitro and in Vivo RNA Inhibition by CD9-HuR Functionalized Exosomes Encapsulated with miRNA or CRISPR/dCas9. Nano Lett. 2019, 19, 19–28. [Google Scholar] [CrossRef]
  110. Porro, C.; Panaro, M.A.; Lofrumento, D.D.; Hasalla, E.; Trotta, T. The multiple roles of exosomes in Parkinson’s disease: An overview. Immunopharmacol. Immunotoxicol. 2019, 41, 469–476. [Google Scholar] [CrossRef]
  111. Vogel, A.D.; Upadhya, R.; Shetty, A.K. Neural stem cell derived extracellular vesicles: Attributes and prospects for treating neurodegenerative disorders. Ebiomedicine 2018, 38, 273–282. [Google Scholar] [CrossRef]
  112. Danzer, K.M.; Kranich, L.R.; Ruf, W.P.; Cagsal-Getkin, O.; Winslow, A.R.; Zhu, L.; Vanderburg, C.R.; McLean, P.J. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 2012, 7, 42. [Google Scholar] [CrossRef] [Green Version]
  113. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  114. Reed, S.L.; Escayg, A. Extracellular vesicles in the treatment of neurological disorders. Neurobiol. Dis. 2021, 157, 105445. [Google Scholar] [CrossRef]
  115. Wang, N.; Mi, X.; Gao, B.; Gu, J.; Wang, W.; Zhang, Y.; Wang, X. Minocycline inhibits brain inflammation and attenuates spontaneous recurrent seizures following pilocarpine-induced status epilepticus. Neuroscience 2015, 287, 144–156. [Google Scholar] [CrossRef]
  116. Ding, M.; Shen, Y.; Wang, P.; Xie, Z.; Xu, S.; Zhu, Z.; Wang, Y.; Lyu, Y.; Wang, D.; Xu, L.; et al. Exosomes Isolated from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Neuroinflammation and Reduce Amyloid-Beta Deposition by Modulating Microglial Activation in Alzheimer’s Disease. Neurochem. Res. 2018, 43, 2165–2177. [Google Scholar] [CrossRef]
  117. Calabria, E.; Scambi, I.; Bonafede, R.; Schiaffino, L.; Peroni, D.; Potrich, V.; Capelli, C.; Schena, F.; Mariotti, R. ASCs-Exosomes Recover Coupling Efficiency and Mitochondrial Membrane Potential in an in vitro Model of ALS. Front. Neurosci. 2019, 13, 1070. [Google Scholar] [CrossRef] [Green Version]
  118. Yang, L.; Zhai, Y.; Hao, Y.; Zhu, Z.; Cheng, G. The Regulatory Functionality of Exosomes Derived from hUMSCs in 3D Culture for Alzheimer’s Disease Therapy. Small 2020, 16, e1906273. [Google Scholar] [CrossRef]
  119. Riazifar, M.; Pone, E.J.; Lötvall, J.; Zhao, W. Stem Cell Extracellular Vesicles: Extended Messages of Regeneration. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 125–154. [Google Scholar] [CrossRef] [Green Version]
  120. Mead, B.; Tomarev, S. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. Stem. Cells Transl. Med. 2017, 6, 1273–1285. [Google Scholar] [CrossRef]
  121. Elia, C.A.; Tamborini, M.; Rasile, M.; Desiato, G.; Marchetti, S.; Swuec, P.; Mazzitelli, S.; Clemente, F.; Anselmo, A.; Matteoli, M.; et al. Intracerebral Injection of Extracellular Vesicles from Mesenchymal Stem Cells Exerts Reduced Aβ Plaque Burden in Early Stages of a Preclinical Model of Alzheimer’s Disease. Cells 2019, 8, 1059. [Google Scholar] [CrossRef] [Green Version]
  122. Canales-Aguirre, A.; Reza-Zaldivar, E.; Hernández-Sapiéns, M.; Gutiérrez-Mercado, Y.K.; Sandoval-Ávila, S.; Gomez-Pinedo, U.; Márquez-Aguirre, A.L.; Vázquez-Méndez, E.; Padilla-Camberos, E. Mesenchymal stem cell-derived exosomes promote neurogenesis and cognitive function recovery in a mouse model of Alzheimer’s disease. Neural Regen. Res. 2019, 14, 1626–1634. [Google Scholar] [CrossRef]
  123. Chen, C.C.; Liu, L.; Ma, F.; Wong, C.W.; Guo, X.E.; Chacko, J.V.; Farhoodi, H.P.; Zhang, S.X.; Zimak, J.; Ségaliny, A.; et al. Elucidation of Exosome Migration Across the Blood–Brain Barrier Model In Vitro. Cell. Mol. Bioeng. 2016, 9, 509–529. [Google Scholar] [CrossRef] [Green Version]
  124. Liu, S.; Fan, M.; Xu, J.-X.; Yang, L.-J.; Qi, C.-C.; Xia, Q.-R.; Ge, J.-F. Exosomes derived from bone-marrow mesenchymal stem cells alleviate cognitive decline in AD-like mice by improving BDNF-related neuropathology. J. Neuroinflamm. 2022, 19, 35. [Google Scholar] [CrossRef]
  125. Kwok, Z.H.; Wang, C.; Jin, Y. Extracellular Vesicle Transportation and Uptake by Recipient Cells: A Critical Process to Regulate Human Diseases. Processes 2021, 9, 273. [Google Scholar] [CrossRef] [PubMed]
  126. Jurgielewicz, B.; Yao, Y.; Stice, S.L. Kinetics and Specificity of HEK293T Extracellular Vesicle Uptake using Imaging Flow Cytometry. Nanoscale Res. Lett. 2020, 15, 170. [Google Scholar] [CrossRef] [PubMed]
  127. Rana, S.; Yue, S.; Stadel, D.; Zöller, M. Toward tailored exosomes: The exosomal tetraspanin web contributes to target cell selection. Int. J. Biochem. Cell Biol. 2012, 44, 1574–1584. [Google Scholar] [CrossRef] [PubMed]
  128. Gómez-Molina, C.; Sandoval, M.; Henzi, R.; Ramírez, J.P.; Varas-Godoy, M.; Luarte, A.; Lafourcade, C.A.; Lopez-Verrilli, A.; Smalla, K.-H.; Kaehne, T.; et al. Small Extracellular Vesicles in Rat Serum Contain Astrocyte-Derived Protein Biomarkers of Repetitive Stress. Int. J. Neuropsychopharmacol. 2019, 22, 232–246. [Google Scholar] [CrossRef] [PubMed]
  129. Chen, H.; Wang, L.; Zeng, X.; Schwarz, H.; Nanda, H.S.; Peng, X.; Zhou, Y. Exosomes, a New Star for Targeted Delivery. Front. Cell Dev. Biol. 2021, 9, 751079. [Google Scholar] [CrossRef] [PubMed]
  130. Yamashita, T.; Takahashi, Y.; Takakura, Y. Possibility of Exosome-Based Therapeutics and Challenges in Production of Exosomes Eligible for Therapeutic Application. Biol. Pharm. Bull. 2018, 41, 835–842. [Google Scholar] [CrossRef] [Green Version]
  131. Jarmalavičiūtė, A.; Tunaitis, V.; Pivoraitė, U.; Venalis, A.; Pivoriūnas, A. Exosomes from dental pulp stem cells rescue human dopaminergic neurons from 6-hydroxy-dopamine–induced apoptosis. Cytotherapy 2015, 17, 932–939. [Google Scholar] [CrossRef]
  132. Fernandes, A.; Ribeiro, A.R.; Monteiro, M.; Garcia, G.; Vaz, A.R.; Brites, D. Secretome from SH-SY5Y APPSwe cells trigger time-dependent CHME3 microglia activation phenotypes, ultimately leading to miR-21 exosome shuttling. Biochimie 2018, 155, 67–82. [Google Scholar] [CrossRef]
  133. Loch-Neckel, G.; Matos, A.T.; Vaz, A.R.; Brites, D. Challenges in the Development of Drug Delivery Systems Based on Small Extracellular Vesicles for Therapy of Brain Diseases. Front. Pharmacol. 2022, 13, 839790. [Google Scholar] [CrossRef]
  134. Strimpakos, A.; Sharma, R.A. Curcumin: Preventive and Therapeutic Properties in Laboratory Studies and Clinical Trials. Antioxid. Redox Signal. 2008, 10, 511–546. [Google Scholar] [CrossRef]
  135. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.-G. A Novel Nanoparticle Drug Delivery System: The Anti-inflammatory Activity of Curcumin Is Enhanced When Encapsulated in Exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef]
  136. Hu, S.; Maiti, P.; Ma, Q.; Zuo, X.; Jones, M.R.; Cole, G.M.; Frautschy, S.A. Clinical development of curcumin in neurodegenerative disease. Expert Rev. Neurother. 2015, 15, 629–637. [Google Scholar] [CrossRef]
  137. Qu, M.; Lin, Q.; Huang, L.; Fu, Y.; Wang, L.; He, S.; Fu, Y.; Yang, S.; Zhang, Z.; Zhang, L.; et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J. Control. Release 2018, 287, 156–166. [Google Scholar] [CrossRef]
  138. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [Green Version]
  139. Zhao, Y.; Haney, M.J.; Gupta, R.; Bohnsack, J.P.; He, Z.; Kabanov, A.V.; Batrakova, E.V. GDNF-Transfected Macrophages Produce Potent Neuroprotective Effects in Parkinson’s Disease Mouse Model. PLoS ONE 2014, 9, e106867. [Google Scholar] [CrossRef] [Green Version]
  140. Lai, R.C.; Yeo, R.W.Y.; Tan, K.H.; Lim, S.K. Exosomes for drug delivery—A novel application for the mesenchymal stem cell. Biotechnol. Adv. 2013, 31, 543–551. [Google Scholar] [CrossRef]
  141. Haney, M.J.; Zhao, Y.; Fallon, J.K.; Yue, W.; Li, S.M.; Lentz, E.E.; Erie, D.; Smith, P.C.; Batrakova, E.V. Extracellular Vesicles as Drug Delivery System for the Treatment of Neurodegenerative Disorders: Optimization of the Cell Source. Adv. NanoBiomed Res. 2021, 1, 2100064. [Google Scholar] [CrossRef]
  142. Henning-Knechtel, A.; Kumar, S.; Wallin, C.; Król, S.; Wärmländer, S.K.; Jarvet, J.; Esposito, G.; Kirmizialtin, S.; Gräslund, A.; Hamilton, A.D.; et al. Designed Cell-Penetrating Peptide Inhibitors of Amyloid-beta Aggregation and Cytotoxicity. Cell Rep. Phys. Sci. 2020, 1, 100014. [Google Scholar] [CrossRef]
  143. Dietz, G.P.H.; Bähr, M. Synthesis of cell-penetrating peptides and their application in neurobiology. Methods Mol. Biol. 2007, 399, 181–198. [Google Scholar]
  144. Magzoub, M.; Gräslund, A. Cell-penetrating peptides: [corrected] from inception to application. Q. Rev. Biophys. 2004, 37, 147–195. [Google Scholar] [CrossRef] [PubMed]
  145. Ruseska, I.; Zimmer, A. Internalization mechanisms of cell-penetrating peptides. Beilstein J. Nanotechnol. 2020, 11, 101–123. [Google Scholar] [CrossRef] [PubMed]
  146. Madani, F.; Lindberg, S.; Langel, Ü.; Futaki, S.; Gräslund, A. Mechanisms of Cellular Uptake of Cell-Penetrating Peptides. J. Biophys. 2011, 2011, 414729. [Google Scholar] [CrossRef] [Green Version]
  147. Langel, Ü. Cell-Penetrating Peptides and Transportan. Pharmaceutics 2021, 13, 987. [Google Scholar] [CrossRef] [PubMed]
  148. Gerbal-Chaloin, S.; Gondeau, C.; Aldrian-Herrada, G.; Heitz, F.; Gauthier-Rouviere, C.; Divita, G. First step of the cell-penetrating peptide mechanism involves Rac1 GTPase-dependent actin-network remodelling. Biol. Cell 2007, 99, 223–238. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, Z.Q.; Hölscher, C. GIP has neuroprotective effects in Alzheimer and Parkinson’s disease models. Peptides 2020, 125, 170184. [Google Scholar] [CrossRef] [PubMed]
  150. Athauda, D.; Foltynie, T. The glucagon-like peptide 1 (GLP) receptor as a therapeutic target in Parkinson’s disease: Mechanisms of action. Drug Discov. Today 2016, 21, 802–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Hölscher, C. Protective properties of GLP-1 and associated peptide hormones in neurodegenerative disorders. Br. J. Pharmacol. 2022, 179, 695–714. [Google Scholar] [CrossRef]
  152. Magzoub, M. Combating Proteins with Proteins: Engineering Cell-Penetrating Peptide Antagonists of Amyloid-β Aggregation and Associated Neurotoxicity. DNA Cell Biol. 2020, 39, 920–925. [Google Scholar] [CrossRef]
  153. Liu, M.; Fang, X.; Yang, Y.; Wang, C. Peptide-Enabled Targeted Delivery Systems for Therapeutic Applications. Front. Bioeng. Biotechnol. 2021, 9, 701504. [Google Scholar] [CrossRef]
  154. Foged, C.; Nielsen, H.M. Cell-penetrating peptides for drug delivery across membrane barriers. Expert Opin. Drug Deliv. 2008, 5, 105–117. [Google Scholar] [CrossRef]
  155. El-Andaloussi, S.; Järver, P.; Johansson, H.J.; Langel, U. Cargo-dependent cytotoxicity and delivery efficacy of cell-penetrating peptides: A comparative study. Biochem. J. 2007, 407, 285–292. [Google Scholar] [CrossRef] [Green Version]
  156. Singh, T.; Murthy, A.S.N.; Yang, H.-J.; Im, J. Versatility of cell-penetrating peptides for intracellular delivery of siRNA. Drug Deliv. 2018, 25, 1996–2006. [Google Scholar] [CrossRef] [Green Version]
  157. Kardani, K.; Milani, A.; Shabani, S.H.; Bolhassani, A. Cell penetrating peptides: The potent multi-cargo intracellular carriers. Expert Opin. Drug Deliv. 2019, 16, 1227–1258. [Google Scholar] [CrossRef]
  158. Fonseca, S.B.; Pereira, M.P.; Kelley, S.O. Recent advances in the use of cell-penetrating peptides for medical and biological applications. Adv. Drug Deliv. Rev. 2009, 61, 953–964. [Google Scholar] [CrossRef]
  159. Ramsey, J.D.; Flynn, N.H. Cell-penetrating peptides transport therapeutics into cells. Pharmacol. Ther. 2015, 154, 78–86. [Google Scholar] [CrossRef] [Green Version]
  160. Li, H.; Tsui, T.Y.; Ma, W. Intracellular Delivery of Molecular Cargo Using Cell-Penetrating Peptides and the Combination Strategies. Int. J. Mol. Sci. 2015, 16, 19518–19536. [Google Scholar] [CrossRef] [Green Version]
  161. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  162. Wightman, B.; Ha, I.; Ruvkun, G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993, 75, 855–862. [Google Scholar] [CrossRef]
  163. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  164. Friedländer, M.R.; Lizano, E.; Houben, A.J.S.; Bezdan, D.; Báñez-Coronel, M.; Kudla, G.; Mateu-Huertas, E.; Kagerbauer, B.; González, J.; Chen, K.C.; et al. Evidence for the biogenesis of more than 1,000 novel human microRNAs. Genome Biol. 2014, 15, R57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Li, S.-C.; Chan, W.-C.; Hu, L.-Y.; Lai, C.-H.; Hsu, C.-N.; Lin, W.-C. Identification of homologous microRNAs in 56 animal genomes. Genomics 2010, 96, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Davis-Dusenbery, B.N.; Hata, A. Mechanisms of control of microRNA biogenesis. J. Biochem. 2010, 148, 381–392. [Google Scholar]
  167. Pasquinelli, A.E.; Reinhart, B.J.; Slack, F.; Martindale, M.Q.; Kuroda, M.I.; Maller, B.; Hayward, D.C.; Ball, E.E.; Degnan, B.; Müller, P.; et al. Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 2000, 408, 86–89. [Google Scholar] [CrossRef] [PubMed]
  168. Broughton, J.P.; Lovci, M.T.; Huang, J.L.; Yeo, G.W.; Pasquinelli, A.E. Pairing beyond the Seed Supports MicroRNA Targeting Specificity. Mol. Cell 2016, 64, 320–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Coller, H.A.; Sang, L.; Roberts, J.M. A New Description of Cellular Quiescence. PLoS Biol. 2006, 4, e83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Maciotta, S.; Meregalli, M.; Torrente, Y. The involvement of microRNAs in neurodegenerative diseases. Front. Cell Neurosci. 2013, 7, 265. [Google Scholar] [CrossRef] [Green Version]
  171. Friedman, R.C.; Farh, K.K.-H.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef] [Green Version]
  172. Sadlon, A.; Takousis, P.; Alexopoulos, P.; Evangelou, E.; Prokopenko, I.; Perneczky, R. miRNAs Identify Shared Pathways in Alzheimer’s and Parkinson’s Diseases. Trends Mol. Med. 2019, 25, 662–672. [Google Scholar] [CrossRef]
  173. Siedlecki-Wullich, D.; Miñano-Molina, A.; Rodríguez-Álvarez, J. microRNAs as Early Biomarkers of Alzheimer’s Disease: A Synaptic Perspective. Cells 2021, 10, 113. [Google Scholar] [CrossRef]
  174. Roser, A.E.; Gomes, L.C.; Schünemann, J.; Maass, F.; Lingor, P. Circulating miRNAs as Diagnostic Biomarkers for Parkinson’s Disease. Front. Neurosci. 2018, 12, 625. [Google Scholar] [CrossRef] [PubMed]
  175. Barbagallo, C.; Mostile, G.; Baglieri, G.; Giunta, F.; Luca, A.; Raciti, L.; Zappia, M.; Purrello, M.; Ragusa, M.; Nicoletti, A. Specific Signatures of Serum miRNAs as Potential Biomarkers to Discriminate Clinically Similar Neurodegenerative and Vascular-Related Diseases. Cell. Mol. Neurobiol. 2020, 40, 531–546. [Google Scholar] [CrossRef] [PubMed]
  176. Kiko, T.; Nakagawa, K.; Tsuduki, T.; Furukawa, K.; Arai, H.; Miyazawa, T. MicroRNAs in Plasma and Cerebrospinal Fluid as Potential Markers for Alzheimer’s Disease. J. Alzheimers Dis. 2014, 39, 253–259. [Google Scholar] [CrossRef] [PubMed]
  177. Schwienbacher, C.; Foco, L.; Picard, A.; Corradi, E.; Serafin, A.; Panzer, J.; Zanigni, S.; Blankenburg, H.; Facheris, M.F.; Giannini, G.; et al. Plasma and White Blood Cells Show Different miRNA Expression Profiles in Parkinson’s Disease. J. Mol. Neurosci. 2017, 62, 244–254. [Google Scholar] [CrossRef] [PubMed]
  178. Ding, H.; Huang, Z.; Chen, M.; Wang, C.; Chen, X.; Chen, J.; Zhang, J. Identification of a panel of five serum miRNAs as a biomarker for Parkinson’s disease. Park. Relat. Disord. 2016, 22, 68–73. [Google Scholar] [CrossRef]
  179. Hou, T.-Y.; Zhou, Y.; Zhu, L.-S.; Wang, X.; Pang, P.; Wang, D.-Q.; Liuyang, Z.-Y.; Man, H.; Lu, Y.; Zhu, L.-Q.; et al. Correcting abnormalities in miR-124/PTPN1 signaling rescues tau pathology in Alzheimer’s disease. J. Neurochem. 2020, 154, 441–457. [Google Scholar] [CrossRef]
  180. Basak, I.; Patil, K.S.; Alves, G.; Larsen, J.P.; Møller, S.G. microRNAs as neuroregulators, biomarkers and therapeutic agents in neurodegenerative diseases. Cell. Mol. Life Sci. 2016, 73, 811–827. [Google Scholar] [CrossRef]
  181. Miñones-Moyano, E.; Porta, S.; Escaramís, G.; Rabionet, R.; Iraola, S.; Kagerbauer, B.; Espinosa-Parrilla, Y.; Ferrer, I.; Estivill, X.; Martí, E. MicroRNA profiling of Parkinson’s disease brains identifies early downregulation of miR-34b/c which modulate mitochondrial function. Hum. Mol. Genet. 2011, 20, 3067–3078. [Google Scholar] [CrossRef] [Green Version]
  182. Esteves, M.; Abreu, R.; Fernandes, H.; Serra-Almeida, C.; Martins, P.A.; Barão, M.; Cristóvão, A.C.; Saraiva, C.; Ferreira, R.; Ferreira, L.; et al. MicroRNA-124-3p-enriched small extracellular vesicles as a therapeutic approach for Parkinson’s disease. Mol. Ther. 2022, 30, 3176–3192. [Google Scholar] [CrossRef]
  183. Recasens, A.; Perier, C.; Sue, C.M. Role of microRNAs in the Regulation of alpha-Synuclein Expression: A Systematic Review. Front. Mol. Neurosci. 2016, 9, 128. [Google Scholar] [CrossRef] [Green Version]
  184. Guy, R.; Offen, D. Promising Opportunities for Treating Neurodegenerative Diseases with Mesenchymal Stem Cell-Derived Exosomes. Biomolecules 2020, 10, 1320. [Google Scholar] [CrossRef] [PubMed]
  185. Elsharkasy, O.; Nordin, J.; Hagey, D.; de Jong, O.; Schiffelers, R.; El Andaloussi, S.; Vader, P. Extracellular vesicles as drug delivery systems: Why and how? Adv. Drug Deliv. Rev. 2020, 159, 332–343. [Google Scholar] [CrossRef] [PubMed]
  186. Pedrioli, G.; Piovesana, E.; Vacchi, E.; Balbi, C. Extracellular Vesicles as Promising Carriers in Drug Delivery: Considerations from a Cell Biologist’s Perspective. Biology 2021, 10, 376. [Google Scholar] [CrossRef] [PubMed]
  187. Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 2021, 16, 748–759. [Google Scholar] [CrossRef] [PubMed]
  188. Lalatsa, A.; Schatzlein, A.G.; Uchegbu, I.F. Strategies to Deliver Peptide Drugs to the Brain. Mol. Pharm. 2014, 11, 1081–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Upadhya, R.; Shetty, A.K. Extracellular Vesicles for the Diagnosis and Treatment of Parkinson’s Disease. Aging Dis. 2021, 12, 1438–1450. [Google Scholar] [CrossRef]
  190. Vandendriessche, C.; Bruggeman, A.; Van Cauwenberghe, C.; Vandenbroucke, R. Extracellular Vesicles in Alzheimer’s and Parkinson’s Disease: Small Entities with Large Consequences. Cells 2020, 9, 2485. [Google Scholar] [CrossRef]
  191. Berumen Sánchez, G.; Bunn, K.E.; Pua, H.H.; Rafat, M. Extracellular vesicles: Mediators of intercellular communication in tissue injury and disease. Cell Commun. Signal 2021, 19, 104. [Google Scholar] [CrossRef]
  192. Coleman, B.M.; Hill, A.F. Extracellular vesicles—Their role in the packaging and spread of misfolded proteins associated with neurodegenerative diseases. Semin. Cell Dev. Biol. 2015, 40, 89–96. [Google Scholar] [CrossRef]
  193. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef]
  194. Hoppstädter, J.; Dembek, A.; Linnenberger, R.; Dahlem, C.; Barghash, A.; Fecher-Trost, C.; Fuhrmann, G.; Koch, M.; Kraegeloh, A.; Huwer, H.; et al. Toll-Like Receptor 2 Release by Macrophages: An Anti-inflammatory Program Induced by Glucocorticoids and Lipopolysaccharide. Front. Immunol. 2019, 10, 1634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Szepesi, Z.; Manouchehrian, O.; Bachiller, S.; Deierborg, T. Bidirectional Microglia–Neuron Communication in Health and Disease. Front. Cell. Neurosci. 2018, 12, 323. [Google Scholar] [CrossRef] [PubMed]
  196. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J.A. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef] [PubMed]
  197. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Delpech, J.-C.; Herron, S.; Botros, M.B.; Ikezu, T. Neuroimmune Crosstalk through Extracellular Vesicles in Health and Disease. Trends Neurosci. 2019, 42, 361–372. [Google Scholar] [CrossRef]
  199. Sun, P.; Liu, D.Z.; Jickling, G.C.; Sharp, F.R.; Yin, K.-J. MicroRNA-based therapeutics in central nervous system injuries. J. Cereb. Blood Flow Metab. 2018, 38, 1125–1148. [Google Scholar] [CrossRef]
  200. Michlewski, G.; Cáceres, J.F. Post-transcriptional control of miRNA biogenesis. RNA 2019, 25, 1–16. [Google Scholar] [CrossRef] [Green Version]
  201. Bernardo, B.C.; Ooi, J.Y.Y.; Lin, R.C.Y.; McMullen, J.R. miRNA therapeutics: A new class of drugs with potential therapeutic applications in the heart. Future Med. Chem. 2015, 7, 1771–1792. [Google Scholar] [CrossRef]
  202. Sonntag, K.-C. MicroRNAs and deregulated gene expression networks in neurodegeneration. Brain Res. 2010, 1338, 48–57. [Google Scholar] [CrossRef] [Green Version]
  203. Faghihi, M.A.; Modarresi, F.; Khalil, A.M.; Wood, D.E.; Sahagan, B.G.; Morgan, T.E.; Finch, C.E.; Laurent, G.S.; Kenny, P.J.; Wahlestedt, C. Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of beta-secretase. Nat. Med. 2008, 14, 723–730. [Google Scholar] [CrossRef] [Green Version]
  204. Cogswell, J.P.; Ward, J.; Taylor, I.A.; Waters, M.; Shi, Y.; Cannon, B.; Kelnar, K.; Kemppainen, J.; Brown, D.; Chen, C.; et al. Identification of miRNA changes in Alzheimer’s disease brain and CSF yields putative biomarkers and insights into disease pathways. J. Alzheimers Dis. 2008, 14, 27–41. [Google Scholar] [CrossRef] [PubMed]
  205. Goh, S.Y.; Chao, Y.X.; Dheen, S.T.; Tan, E.-K.; Tay, S.S.-W. Role of micrornas in parkinson’s disease. Int. J. Mol. Sci. 2019, 20, 5649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Junn, E.; Mouradian, M.M. MicroRNAs in neurodegenerative diseases and their therapeutic potential. Pharmacol. Ther. 2012, 133, 142–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Choi, D.C.; Chae, Y.-J.; Kabaria, S.; Chaudhuri, A.D.; Jain, M.R.; Li, H.; Mouradian, M.M.; Junn, E. MicroRNA-7 Protects against 1-Methyl-4-Phenylpyridinium-Induced Cell Death by Targeting RelA. J. Neurosci. 2014, 34, 12725–12737. [Google Scholar] [CrossRef] [Green Version]
  208. Leggio, L.; Vivarelli, S.; L’Episcopo, F.; Tirolo, C.; Caniglia, S.; Testa, N.; Marchetti, B.; Iraci, N. microRNAs in Parkinson’s Disease: From Pathogenesis to Novel Diagnostic and Therapeutic Approaches. Int. J. Mol. Sci. 2017, 18, 2698. [Google Scholar] [CrossRef] [Green Version]
  209. Wiedrick, J.T.; Phillips, J.I.; Lusardi, T.A.; McFarland, T.J.; Lind, B.; Sandau, U.S.; Harrington, C.A.; Lapidus, J.A.; Galasko, D.R.; Quinn, J.F.; et al. Validation of MicroRNA Biomarkers for Alzheimer’s Disease in Human Cerebrospinal Fluid. J. Alzheimers Dis. 2019, 67, 875–891. [Google Scholar] [CrossRef]
  210. Takousis, P.; Sadlon, A.; Schulz, J.; Wohlers, I.; Dobricic, V.; Middleton, L.; Lill, C.M.; Perneczky, R.; Bertram, L. Differential expression of microRNAs in Alzheimer’s disease brain, blood, and cerebrospinal fluid. Alzheimers Dement. 2019, 15, 1468–1477. [Google Scholar] [CrossRef]
  211. Swarbrick, S.; Wragg, N.; Ghosh, S.; Stolzing, A. Systematic Review of miRNA as Biomarkers in Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 6156–6167. [Google Scholar] [CrossRef] [Green Version]
  212. Keller, A.; Backes, C.; Haas, J.; Leidinger, P.; Maetzler, W.; Deuschle, C.; Berg, D.; Ruschil, C.; Galata, V.; Ruprecht, K.; et al. Validating Alzheimer’s disease micro RNAs using next-generation sequencing. Alzheimers Dement. 2016, 12, 565–576. [Google Scholar] [CrossRef]
  213. Higaki, S.; Muramatsu, M.; Matsuda, A.; Matsumoto, K.; Satoh, J.-I.; Michikawa, M.; Niida, S. Defensive effect of microRNA-200b/c against amyloid-beta peptide-induced toxicity in Alzheimer’s disease models. PLoS ONE 2018, 13, e0196929. [Google Scholar] [CrossRef]
  214. Munir, J.; Yoon, J.K.; Ryu, S. Therapeutic miRNA-Enriched Extracellular Vesicles: Current Approaches and Future Prospects. Cells 2020, 9, 2271. [Google Scholar] [CrossRef] [PubMed]
  215. Berry, C. Intracellular delivery of nanoparticles via the HIV-1 tat peptide. Nanomedicine 2008, 3, 357–365. [Google Scholar] [CrossRef] [PubMed]
  216. Rizzuti, M.; Nizzardo, M.; Zanetta, C.; Ramirez, A.; Corti, S. Therapeutic applications of the cell-penetrating HIV-1 Tat peptide. Drug Discov. Today 2015, 20, 76–85. [Google Scholar] [CrossRef] [PubMed]
  217. Reissmann, S. Cell penetration: Scope and limitations by the application of cell-penetrating peptides. J. Pept. Sci. 2014, 20, 760–784. [Google Scholar] [CrossRef]
  218. Nakase, I.; Noguchi, K.; Aoki, A.; Takatani-Nakase, T.; Fujii, I.; Futaki, S. Arginine-rich cell-penetrating peptide-modified extracellular vesicles for active macropinocytosis induction and efficient intracellular delivery. Sci. Rep. 2017, 7, 1991. [Google Scholar] [CrossRef] [Green Version]
  219. Nakase, I. Biofunctional Peptide-Modified Extracellular Vesicles Enable Effective Intracellular Delivery via the Induction of Macropinocytosis. Processes 2021, 9, 224. [Google Scholar] [CrossRef]
  220. Hirase, S.; Aoki, A.; Hattori, Y.; Morimoto, K.; Noguchi, K.; Fujii, I.; Takatani-Nakase, T.; Futaki, S.; Kirihata, M.; Nakase, I. Dodecaborate-Encapsulated Extracellular Vesicles with Modification of Cell-Penetrating Peptides for Enhancing Macropinocytotic Cellular Uptake and Biological Activity in Boron Neutron Capture Therapy. Mol. Pharm. 2022, 19, 1135–1145. [Google Scholar] [CrossRef]
  221. Mashouri, L.; Yousefi, H.; Aref, A.R.; Ahadi, A.M.; Molaei, F.; Alahari, S.K. Exosomes: Composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol. Cancer 2019, 18, 75. [Google Scholar] [CrossRef] [Green Version]
  222. García-Manrique, P.; Matos, M.; Gutiérrez, G.; Pazos, C.; Blanco-López, M.C. Therapeutic biomaterials based on extracellular vesicles: Classification of bio-engineering and mimetic preparation routes. J. Extracell. Vesicles 2018, 7, 1422676. [Google Scholar] [CrossRef] [Green Version]
  223. Pascucci, L.; Coccè, V.; Bonomi, A.; Ami, D.; Ceccarelli, P.; Ciusani, E.; Viganò, L.; Locatelli, A.; Sisto, F.; Doglia, S.M.; et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: A new approach for drug delivery. J. Control. Release 2014, 192, 262–270. [Google Scholar] [CrossRef]
  224. Li, S.-P.; Lin, Z.-X.; Jiang, X.-Y.; Yu, X.-Y. Exosomal cargo-loading and synthetic exosome-mimics as potential therapeutic tools. Acta Pharmacol. Sin. 2018, 39, 542–551. [Google Scholar] [CrossRef] [Green Version]
  225. van der Meel, R.; Fens, M.H.A.M.; Vader, P.; van Solinge, W.W.; Eniola-Adefeso, O.; Schiffelers, R.M. Extracellular vesicles as drug delivery systems: Lessons from the liposome field. J. Control. Release 2014, 195, 72–85. [Google Scholar] [CrossRef]
  226. Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [Green Version]
  227. Steinman, R.M. Decisions About Dendritic Cells: Past, Present, and Future. Annu. Rev. Immunol. 2012, 30, 1–22. [Google Scholar] [CrossRef] [Green Version]
  228. Li, L.; Lu, S.; Liang, X.; Cao, B.; Wang, S.; Jiang, J.; Luo, H.; He, S.; Lang, J.; Zhu, G. γδTDEs: An Efficient Delivery System for miR-138 with Anti-tumoral and Immunostimulatory Roles on Oral Squamous Cell Carcinoma. Mol. Ther. Nucleic Acids 2019, 14, 101–113. [Google Scholar] [CrossRef] [Green Version]
  229. Lee, J.; Kim, J.; Jeong, M.; Lee, H.; Goh, U.; Kim, H.; Kim, B.; Park, J.-H. Liposome-Based Engineering of Cells to Package Hydrophobic Compounds in Membrane Vesicles for Tumor Penetration. Nano Lett. 2015, 15, 2938–2944. [Google Scholar] [CrossRef] [PubMed]
  230. Cheng, L.; Wang, Y.; Huang, L. Exosomes from M1-Polarized Macrophages Potentiate the Cancer Vaccine by Creating a Pro-inflammatory Microenvironment in the Lymph Node. Mol. Ther. 2017, 25, 1665–1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Pessina, A.; Bonomi, A.; Coccè, V.; Invernici, G.; Navone, S.; Cavicchini, L.; Sisto, F.; Ferrari, M.; Viganò, L.; Locatelli, A.; et al. Mesenchymal Stromal Cells Primed with Paclitaxel Provide a New Approach for Cancer Therapy. PLoS ONE 2011, 6, e28321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Vergauwen, G.; Dhondt, B.; Van Deun, J.; De Smedt, E.; Berx, G.; Timmerman, E.; Gevaert, K.; Miinalainen, I.; Cocquyt, V.; Braems, G.; et al. Confounding factors of ultrafiltration and protein analysis in extracellular vesicle research. Sci. Rep. 2017, 7, 2704. [Google Scholar] [CrossRef] [Green Version]
  234. Stranska, R.; Gysbrechts, L.; Wouters, J.; Vermeersch, P.; Bloch, K.; Dierickx, D.; Andrei, G.; Snoeck, R. Comparison of membrane affinity-based method with size-exclusion chromatography for isolation of exosome-like vesicles from human plasma. J. Transl. Med. 2018, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  235. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Botchway, B.O.; Okoye, F.C.; Chen, Y.; Arthur, W.E.; Fang, M. Alzheimer Disease: Recent Updates on Apolipoprotein E and Gut Microbiome Mediation of Oxidative Stress, and Prospective Interventional Agents. Aging Dis. 2022, 13, 87–102. [Google Scholar] [CrossRef] [PubMed]
  237. Brandebura, A.N.; Paumier, A.; Onur, T.S.; Allen, N.J. Astrocyte contribution to dysfunction, risk and progression in neurodegenerative disorders. Nat. Rev. Neurosci. 2023, 24, 23–39. [Google Scholar] [CrossRef] [PubMed]
  238. Pachler, K.; Lener, T.; Streif, D.; Dunai, Z.A.; Desgeorges, A.; Feichtner, M.; Öller, M.; Schallmoser, K.; Rohde, E.; Gimona, M. A Good Manufacturing Practice–grade standard protocol for exclusively human mesenchymal stromal cell–derived extracellular vesicles. Cytotherapy 2017, 19, 458–472. [Google Scholar] [CrossRef] [Green Version]
  239. Reissmann, S.; Filatova, M.P. New generation of cell-penetrating peptides: Functionality and potential clinical application. J. Pept. Sci. 2021, 27, e3300. [Google Scholar] [CrossRef]
  240. Palm, C.; Jayamanne, M.; Kjellander, M.; Hällbrink, M. Peptide degradation is a critical determinant for cell-penetrating peptide uptake. Biochim. Biophys. Acta 2007, 1768, 1769–1776. [Google Scholar] [CrossRef] [Green Version]
  241. Hirose, H.; Takeuchi, T.; Osakada, H.; Pujals, S.; Katayama, S.; Nakase, I.; Kobayashi, S.; Haraguchi, T.; Futaki, S. Transient Focal Membrane Deformation Induced by Arginine-rich Peptides Leads to Their Direct Penetration into Cells. Mol. Ther. 2012, 20, 984–993. [Google Scholar] [CrossRef] [Green Version]
  242. Seisel, Q.; Pelletier, F.; Deshayes, S.; Boisguerin, P. How to evaluate the cellular uptake of CPPs with fluorescence techniques: Dissecting methodological pitfalls associated to tryptophan-rich peptides. Biochim. Biophys. Acta Biomembr. 2019, 1861, 1533–1545. [Google Scholar] [CrossRef]
  243. Kristensen, M.; Birch, D.; Nielsen, H.M. Applications and Challenges for Use of Cell-Penetrating Peptides as Delivery Vectors for Peptide and Protein Cargos. Int. J. Mol. Sci. 2016, 17, 185. [Google Scholar] [CrossRef] [Green Version]
  244. Rennert, R.; Wespe, C.; Beck-Sickinger, A.G.; Neundorf, I. Developing novel hCT derived cell-penetrating peptides with improved metabolic stability. Biochim. Biophys. Acta 2006, 1758, 347–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Pathogenesis of Alzheimer’s’ disease and Parkinson’s’ disease. AD and PD share common pathways of degeneration. Both neurodegenerative diseases exhibit significant amounts of oxidative stress, neuronal inflammation and degeneration, as well as the build-up of insoluble proteins including β-amyloid and α-synuclein.
Figure 1. Pathogenesis of Alzheimer’s’ disease and Parkinson’s’ disease. AD and PD share common pathways of degeneration. Both neurodegenerative diseases exhibit significant amounts of oxidative stress, neuronal inflammation and degeneration, as well as the build-up of insoluble proteins including β-amyloid and α-synuclein.
Biomedicines 11 00728 g001
Figure 2. Extracellular vesicles in the therapeutics of neurodegeneration. EVs, derived from mesenchymal stromal cells (MSCs), have been studied in many AD and PD models of neurodegeneration. To date, researchers have noted a correlation between EV-based therapies and a reduction in oxidative stress, protein aggregates, pro inflammatory cytokines and degenerating neurons. In addition, they have also noted a correlation between these therapies and an increase in mitochondrial function and anti-inflammatory cytokines.
Figure 2. Extracellular vesicles in the therapeutics of neurodegeneration. EVs, derived from mesenchymal stromal cells (MSCs), have been studied in many AD and PD models of neurodegeneration. To date, researchers have noted a correlation between EV-based therapies and a reduction in oxidative stress, protein aggregates, pro inflammatory cytokines and degenerating neurons. In addition, they have also noted a correlation between these therapies and an increase in mitochondrial function and anti-inflammatory cytokines.
Biomedicines 11 00728 g002
Figure 3. Cell-penetrating peptides as therapeutics for neurodegeneration. CPPs, either by themselves or in conjunction with proteins, siRNAs, drugs, or DNA, are thought to have many neuroprotective features in AD and PD models of neurodegeneration. When studied, CPP-based therapies reduce protein aggregates, neuronal inflammation and mitochondrial dysfunction while also restoring functional synapses.
Figure 3. Cell-penetrating peptides as therapeutics for neurodegeneration. CPPs, either by themselves or in conjunction with proteins, siRNAs, drugs, or DNA, are thought to have many neuroprotective features in AD and PD models of neurodegeneration. When studied, CPP-based therapies reduce protein aggregates, neuronal inflammation and mitochondrial dysfunction while also restoring functional synapses.
Biomedicines 11 00728 g003
Figure 4. A novel approach to neurodegeneration therapeutics.
Figure 4. A novel approach to neurodegeneration therapeutics.
Biomedicines 11 00728 g004
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

Keighron, C.N.; Avazzadeh, S.; Goljanek-Whysall, K.; McDonagh, B.; Howard, L.; Ritter, T.; Quinlan, L.R. Extracellular Vesicles, Cell-Penetrating Peptides and miRNAs as Future Novel Therapeutic Interventions for Parkinson’s and Alzheimer’s Disease. Biomedicines 2023, 11, 728. https://doi.org/10.3390/biomedicines11030728

AMA Style

Keighron CN, Avazzadeh S, Goljanek-Whysall K, McDonagh B, Howard L, Ritter T, Quinlan LR. Extracellular Vesicles, Cell-Penetrating Peptides and miRNAs as Future Novel Therapeutic Interventions for Parkinson’s and Alzheimer’s Disease. Biomedicines. 2023; 11(3):728. https://doi.org/10.3390/biomedicines11030728

Chicago/Turabian Style

Keighron, Cameron Noah, Sahar Avazzadeh, Katarzyna Goljanek-Whysall, Brian McDonagh, Linda Howard, Thomas Ritter, and Leo R. Quinlan. 2023. "Extracellular Vesicles, Cell-Penetrating Peptides and miRNAs as Future Novel Therapeutic Interventions for Parkinson’s and Alzheimer’s Disease" Biomedicines 11, no. 3: 728. https://doi.org/10.3390/biomedicines11030728

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop