Next Article in Journal
Enhancement and Limitations of Green-Spectrum Dual-Wavelength Irradiation in Porphyrin-Based Antimicrobial Strategies Targeting Cutibacterium acnes subsp. elongatum
Previous Article in Journal
Liraglutide and Exenatide in Alzheimer’s Disease and Mild Cognitive Impairment: A Systematic Review and Meta-Analysis of Cognitive Outcomes
Previous Article in Special Issue
Harnessing Extracellular Vesicles for Targeted Drug Delivery in Ovarian Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extracellular Vesicles in Alzheimer’s Disease: Dual Roles in Pathogenesis, Promising Avenues for Diagnosis and Therapy

by
Feng Li
1,2,3,
Liyang Wu
2,3,
Xin Feng
2,4,
Yihong Li
1,2,3,* and
Huadong Fan
1,2,4,*
1
College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo 315000, China
2
Innovation Center for Diagnosis and Treatment of Neurological Disease, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
3
Lab of Nanopharmacology Research for Neurodegeneration, Department of Research and Development of Science and Technology, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
4
Lab of Dementia and Neurorehabilitation Research, Department of Research Development of Science and Technology, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315000, China
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2026, 18(1), 70; https://doi.org/10.3390/pharmaceutics18010070
Submission received: 10 December 2025 / Revised: 31 December 2025 / Accepted: 3 January 2026 / Published: 5 January 2026

Abstract

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) plaques, neurofibrillary tau tangles, chronic neuroinflammation, and synaptic loss, leading to cognitive decline. Extracellular vesicles (EVs)—lipid bilayer nanoparticles secreted by nearly all cell types—have emerged as critical mediators of intercellular communication, playing a complex dual role in both the pathogenesis and potential treatment of AD. This review generally delineates two opposite roles of EVs in pathogenesis and potential treatment of AD. On one hand, EVs derived from neurons, astrocytes, microglia and oligodendrocytes can propagate toxic proteins (Aβ, tau) and inflammatory signals, thereby accelerating disease progression. On the other hand, EVs—especially those from mesenchymal stem cells (MSCs)—exert neuroprotective effects by facilitating toxic protein clearance, modulating immune responses, preserving synaptic integrity, and alleviating oxidative stress. The cargo-carrying function of EVs gives them considerable diagnostic value. The associated cargos such as proteins and microRNAs (miRNAs) in the EVs may serve as minimally invasive biomarkers for early detection and monitoring of AD. Therapeutically, engineered EVs, including those incorporating CRISPR/Cas9-based genetic modification, are being developed as sophisticated delivery platforms for targeting core AD pathologies. Furthermore, this review highlights emerging technologies such as microfluidic chips and focused ultrasound (FUS), discussing their potential to enhance the translational prospects of EV-based early diagnostic and treatment for AD.

1. Introduction

Alzheimer’s disease (AD) is the most common cause of dementia, affecting over 55 million people worldwide. The number is expected to rise to nearly 152.8 million by 2050, with the most significant increase anticipated in developing countries [1,2]. Pathologically, AD is characterized by the accumulation of misfolded proteins, including amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles, accompanied by neuroinflammation, synaptic dysfunction, and metabolic disturbances [3]. These interconnected processes ultimately lead to progressive neuronal loss and cognitive decline [4,5]. Despite decades of intensive research, effective therapies remain elusive, as most treatments targeting single pathological factors have yielded limited clinical benefits [6].
The pathology of AD is characterized by three main features: (1) The accumulation of Aβ, considered a primary pathogenic driver [7]. Aβ is generated from the amyloid precursor protein (APP) through cleavage by β-secretase (BACE-1) and γ-secretase. The resulting Aβ monomers then aggregate into soluble oligomers, protofibrils and fibrils, ultimately forming amyloid plaques in the brain parenchyma and cerebral blood vessels [8,9]. (2) Neurofibrillary tangles (NFTs), another pathological hallmark, arise from hyper-phosphorylated and misfolded Tau proteins [3,7]. This abnormal phosphorylation disrupts tau’s association with microtubes, causing it to aggregate into NFTs within neurons and impair axonal transport and synaptic function [3]. (3) Increasing evidence identifies neuroinflammation as a critical driver and amplifier of AD pathology [10]. Activated microglia, often clustered around amyloid plaques, respond to pathogenic signals such as Aβ oligomers and other damage-associated molecules. This activation triggers the release of pro-inflammatory cytokines (e.g., IL-1β, TNF-α), chemokines, and reactive oxygen species (ROS), which further exacerbate neuronal dysfunction and synaptic damage [11]. While initially protective, sustained inflammatory responses contributes to neuronal degeneration, mitochondrial dysfunction, synaptic loss and further propagation of both Aβ and tau pathologies [9,12]. Crucially, these pathogenic factors interact synergistically to promote AD pathology. For instance, Aβ oligomers can accelerate tau hyperphosphorylation, while tau pathology facilitates the spread of neurodegeneration across the brain region [13,14].
Given their roles in mediating complex interplay among neurons, glia, and the extracellular environment, extracellular vesicles (EVs) are increasingly recognized as key participants in AD processes, facilitating both the propagation and regulation of Aβ, tau, and inflammatory signaling. EVs are nanoscale, lipid bilayer-enclosed particles secreted by nearly all cell types and are present in biological fluids [15,16,17]. Based on their biogenesis, EVs are broadly classified into exosomes, microvesicles, and apoptotic bodies [18]. Exosomes (30–150 nm) originate from the inward budding of endosomal membranes, forming multivesicular bodies (MVBs) that release intraluminal vesicles (ILVs) into the extracellular space upon fusion with the plasma membrane [19,20]. They are commonly characterized by surface markers such as CD9, CD63, CD81, ALIX (PDCD6IP), and TSG101 [21]. In contrast, microvesicles (100–1000 nm) are generated through direct outward budding and fission of the plasma membrane and are commonly enriched in CD40, selectins and integrins [22,23,24]. Apoptotic bodies (1–5 μm) are released from cells undergoing programmed cell death and typically display markers such as Annexin V [25,26]. Despite differences in size and biogenesis, all EV subtypes transport diverse molecular cargo-including proteins, lipids, nucleic acids (mRNA, miRNA), and metabolites from their parent cells—thereby facilitating intercellular communication and regulating a broad range of physiological and pathological processes [22,27,28] (Table 1).
In the context of AD, EVs exhibited a paradoxical, dual role. On one hand, they propagate disease by transferring toxic proteins such as Aβ or tau, promoting neuroinflammation and accelerating neurodegeneration [29,30]. On the other hand, EVs also exert neuroprotective effects by delivering regulatory molecules that support neuronal survival, modulate immune responses and promote repair mechanisms [31,32]. This bidirectional influence highlights their importance as both mediators and modulators of AD pathology.
Accordingly, this review provides a comprehensive overview of the multifaceted role of EVs in AD. We first discuss the classification and cellular origin of EVs subtypes involved in AD, followed by their involvement in disease pathogenesis and endogenous protective mechanisms. Subsequently, we evaluate how EVs can be harnessed for therapeutic and diagnostic applications. Finally, we explore recent advances, translational challenges, and future perspectives in leveraging EVs for the treatment and early detection of AD.
Table 1. The characteristics and functions of EV subtypes.
Table 1. The characteristics and functions of EV subtypes.
EV SubtypeSize RangeKey Surface MarkersTypical CargoesPrimary FunctionsReferences
Exosomes30–150 nmTetraspanins (CD9, CD63, CD81), ALIX, TSG101, HSP70Proteins, lipids, mRNAs, miRNAs, other non-coding RNAsIntercellular signaling, immune modulation, synaptic plasticity. In AD, heavily implicated in Aβ/tau propagation and neuroinflammation.[33,34]
Microvesicles100–1000 nmIntegrins, selectins, CD40, ARF6, tissue factorCytosolic proteins, lipids, RNAs, organellesIntercellular communication, Stimulate synaptic activity.[24,35]
Apoptotic Bodies1–5 μmPhosphatidylserine (exposed), Annexin V, calreticulin, calnexinPlasma membrane, fragmented organelles, various biomolecules (RNA and DNA)Clearance of apoptotic debris; intercellular communication.[21,26]
NDEVs144–230 nmL1CAM, GluR2/3, GAP43, NLGN3Synaptic proteins, lipids, regulatory miRNAs (e.g., miR-21-5p), and pathogenic molecules (tau and Aβ)Propagation of pathological proteins, participating in synaptic communication and plasticity.[36,37,38]
ADEVs100–160 nmEAAT1/GLAST, GFAPPathogenic molecules (tau and Aβ), PAR-4 and ceramide, heat shock proteins and NTPDasesPromote neurite outgrowth, dendritic branching, neuronal survival, and synaptic plasticity, impair neuronal excitability and neurite growth.[39,40,41]
MDEVs98 ± 24.1 nmCD9, CD81, CD63, MHC class II moleculesPathogenic molecules (tau and Aβ), TRME2, Inflammatory factors (IL-1β and TNF-α), synaptogenic proteinsPropagation of Aß and Tau,
release of inflammatory factors,
synaptic dysfunction, alleviation of oxidative stress, neurite outgrowth.
[42,43,44]
ODEVs30–80 nmALIX, TSG101, Myelin-specific proteins (PLP, CNO, MBP, MOG)PLP, CNP, MBP, MOG, and characteristic myelin lipidsTrigger of neuronal hyperactivity, alleviation of oxidative stress
synaptic preservation.
[45,46,47,48]
MSC-EVs40–100 nmCD9, ALIX, TSG101, CD90, CD73, and CD44Growth factors (e.g., VEGF, TGF-β, HGF), immunomodulatory molecules (IL-10), lipids, and microRNAs (e.g., miR-21, miR-122, miR-146a)Clearance of Aß and Tau,
alleviation of oxidative stress,
synaptic preservation, reduction of neuroinflammation, metabolic modulation and folate delivery.
[49,50,51,52]

2. Classification and Functional Heterogeneity

Beyond structural classification, the biological functions of EVs are primarily determined by the identity and physiological state of their parent cells [53]. The cellular origin governs the specific molecular composition of EV cargo, shaping their effects on recipient cells [54]. The principle is particularly significant within the complex microenvironment of the central nervous system (CNS), where the distinct cargo of EVs derived from different neural cell types—including neurons, astrocytes, microglia, oligodendrocytes, and mesenchymal stem cells—exert distinct and sometimes opposing influences on neural function and disease progression [55,56]. In AD, this cellular heterogeneity underlies the dual role of EVs: while some populations propagate pathological factors such as Aβ and tau, amplify neuroinflammation, and disseminate oxidative stress, others mediate the clearance of toxic proteins and maintain redox homeostasis, thereby protecting against neurodegeneration. Understanding this diversity is therefore crucial for deciphering disease mechanisms and harnessing EVs for therapeutic applications.

2.1. Neuron-Derived Extracellular Vesicles (NDEVs)

NDEVs are characterized by surface markers such as the L1 cell adhesion molecule (L1CAM), GluR2/3, GAP43 and NLGN3 [57,58]. These markers are not only enable the immunocapture and isolation of NDEVs from biofluids but also position them as promising candidates for liquid biopsy in AD [36]. The cargo of NDEVs mirrors neuronal physiology and pathology, containing a diverse array of neuron-specific molecules, includes synaptic proteins, lipids, regulatory miRNAs (e.g., miR-21-5p), and pathogenic molecules such as hyperphosphorylated tau and Aβ. Notably, although numerous studies suggest that NDEVs contribute to AD progression by facilitating the spread of Aβ and tau pathology, other reports indicate that NDEVs may also participate in synaptic maintenance and neuronal stress responses [37,59]. These seemingly conflicting observations likely depend on disease stage, neuronal metabolic status, and EV cargo composition, highlighting unresolved questions regarding the net impact of NDEVs in vivo.

2.2. Astrocyte-Derived Extracellular Vesicles (ADEVs)

ADEVs are identified by specific markers such as excitatory amino acid transporter 1 (EAAT1/GLAST) and glial fibrillary acidic protein (GFAP) [40,60]. In AD, their functional profile is highly context-dependent [61]. Under anti-inflammatory conditions such as ATP or IL-10 stimulation, ADEVs carry neurotropic proteins that promote neurite outgrowth, dendritic branching, neuronal survival, and synaptic plasticity [41,62,63]. In contrast, astrocytes activated by pro-inflammatory cytokines (e.g., IL-1β) release ADEVs enriched in inflammatory mediators that impair neuronal excitability and neurite growth [64,65]. This duality underscores the need for further studies to clarify how astrocyte activation states and microenvironmental cues shape EV function during different stages of AD.

2.3. Microglia-Derived Extracellular Vesicles (MDEVs)

MDEVs originate from the brain’s resident immune cells and are characterized by common exosomal tetraspanin markers (CD9, CD81, and CD63) [66,67]. Notably, MDEVs also express functional major histocompatibility complex (MHC) class II molecules, reflecting their immunological activity [43]. The cargo composition of MDEVs is closely linked to the activation state of their parent microglia, determining whether their function is neuroprotective or neurotoxic [43]. MDEVs derived from pro-inflammatory (M1) microglia promotes neuroinflammation and neuronal injury, while those from alternatively activated (M2) microglia carry anti-inflammatory cytokines and neuroprotective molecules that suppress inflammation and oxidative stress [67,68]. Despite substantial evidence implicating MDEVs in the amplification of neuroinflammation and tau propagation, other studies suggest that MDEVs can also facilitate debris clearance and resolution of inflammation. Such divergent findings likely reflect differences in microglial polarization states, experimental models, and disease progression, emphasizing unresolved questions regarding when MDEVs act as pathogenic mediators versus modulators of tissue repair.

2.4. Oligodendrocyte-Derived Extracellular Vesicles (ODEVs)

ODEVs are characterized by typical exosomal markers, such as ALIX, TSG101, as well as myelin-specific proteins. These include proteolipid protein (PLP), 2′,3′-cyclic nucleotide 3′-phosphodiesterase (CNP), myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), and characteristic myelin lipids [45,46,69]. As essential mediators of oligodendrocyte-axon communication, ODEVs have been shown to be internalized by neurons via endocytosis. Functionally, they support axonal maintenance and metabolic homeostasis [47,70,71]. However, under pathological conditions, ODEVs may also contribute to disease processes, underscoring their complex and context-dependent roles in AD [47]. Notably, compared with neuronal and glial derived EVs, the involvement of ODEVs in AD remains poorly understood. Particularly, the oligodendrocytes are the primary cells for myelin sheath formation, which enables high-speed transmission of electrical signals between neurons [72]. However, whether extracellular vesicles from these cells (ODEVs) influence axonal conductance or regeneration during AD development remains unexplored.

2.5. Mesenchymal Stem Cell-Derived Extracellular Vesicles (MSC-EVs)

MSC-EVs are important paracrine effector molecules of mesenchymal stem cells (MSCs) and have shown therapeutic potential in diverse inflammatory and degenerative diseases [50,73]. These vesicles express universal EV markers such as CD9, ALIX, and TSG101 along with MSC-specific markers, such as CD90, CD73, and CD44 [51,74]. Their cargo includes growth factors (e.g., VEGF, TGF-β, HGF) [75,76], immunomodulatory molecules (IL-10), lipids, and microRNAs (e.g., miR-21, miR-122, miR-146a) [77,78,79]. Importantly, MSC-EVs are not a homogeneous population. Lai and colleagues were the first to demonstrate that MSCs secrete at least three distinct subtypes of EVs with differing membrane lipids, proteins and RNA cargoes, reflecting their diverse biosynthetic origins and specialized biological functions [80]. This intrinsic heterogeneity underpins their versatility as therapeutic candidates for neurodegenerative disorders, including AD.

3. The Dual Role of EVs in Mediating AD Pathogenesis

As established in the previous section, the functional heterogeneity of EVs is intrinsically linked to the physiological state of their parent cells. This context-dependent nature determines its complex dual role in AD, wherein they can function as both drivers of pathological progression and mediators of endogenous protection. This section delineates these opposing functions, first examining the mechanisms by which EVs exacerbate core AD pathologies; namely, the propagation of Aβ and tau, the amplification of neuroinflammation, and the dissemination of oxidative stress. Subsequently, we explore the protective countermeasures orchestrated by EVs, including the clearance of toxic proteins, neuroprotection, immunomodulation, and the preservation of synaptic integrity and metabolic homeostasis (Figure 1).

3.1. The Dark Side: EVs as Mediators of AD Progression

3.1.1. Propagation of Aβ and Tau

A key mechanism by which EVs promote AD progression involves dissemination of toxic proteins—particularly Aβ and hyperphosphorylated tau—across neuronal networks. NDEVs serve as vehicles for these pathogenic proteins [81]. They encapsulate these pathological proteins and release them into the extracellular space, these vesicles are subsequently internalized by neighboring neurons, propagating pathological tau phosphorylation and Aβ accumulation throughout interconnected brain regions [37]. Clinical evidence supports this mechanism: plasma NDEVs from AD patients contain significantly elevated levels of P-tau T181, P-tau S396, and Aβ1–42. Crucially, when these human-derived NDEVs are injected into mice, they induced tau pathology in the hippocampus, directly demonstrating their capacity to propagate neuropathology [38].
Beyond NDEVs, glia-derived EVs—particularly those from astrocytes and microglia—further amplify disease pathology [82,83]. ADEVs contain high levels of APP-processing components, including BACE-1, γ-secretase, soluble Aβ42, and soluble amyloid precursor protein β (sAPPβ), when compared with NDEVs from the same individuals [41,65]. Through this cargo, ADEVs can promote APP cleavage and intercellular transport of amyloidogenic peptides, thereby enhancing local Aβ accumulation. Furthermore, ADEVs also carry pathogenic phosphorylated tau species (p-tau T181 and p-tau S396) [84]. Similarly, MDEVs can encapsulate and release Aβ and tau, which can be transferred to neurons and other glial cells, perpetuating the pathogenic cycle [85]. Emerging evidence further implicates ODEVs may also contribute to AD pathogenesis. Oligodendrocytes have been shown to produce Aβ and to drive abnormal neuronal hyperactivity in AD model [86]. Selective suppression of oligodendrocyte-derived Aβ production rescues neuronal dysfunction and reduces plaque formation, suggesting that ODEVs may act as additional vehicle for delivering Aβ to neurons, thereby promoting neuronal dysfunction and accelerating AD pathology [87]. Collectively, these findings indicate that EVs derived from multiple neural cell types form an interconnected network that drives the intercellular spread of Aβ and tau, thereby perpetuating neurodegeneration and neuroinflammation in AD.

3.1.2. Amplification of Neuroinflammation

The pathogenic role of EVs in AD extends beyond the transmission of misfolded proteins to include the modulation of neuroinflammatory signaling [88]. In this process, microglia and astrocytes, along with their secreted EVs, create a vicious inflammatory cycle that exacerbates neuronal damage. Upon pro-inflammatory activation, microglia release MDEVs loaded with cytokines such as IL-1β and TNF-α, these vesicles further propagate tau pathology by internalizing and subsequently secreting pathological tau proteins [44]. Damage-associated molecules like ATP can trigger the release of such cytokine-enriched MDEVs, which in turn intensify neuroinflammatory signaling in the brain [89,90]. Similarly, astrocytes exposed to IL-1β or Aβ release ADEVs carrying pro-inflammatory and neurotoxic cargo, including PAR-4 and ceramide, which downregulate anti-inflammatory gene expression in recipient cells and further induce neuron apoptosis [61,91]. Functionally, such ADEVs have been shown to impair neurite outgrowth, reduce neuronal excitability, and further stimulate neuronal Aβ production [61]. Together, these interactions establish a pathological feed-forward loop: neuroinflammation recruits and activates more glial cells, which in turn release more inflammatory EVs (both MDEVs and ADEVs), thereby worsening synaptic dysfunction, impairing neuronal communication, and accelerating neurodegeneration [92,93]. Furthermore, under AD-related stress, the typically homeostatic ODEVs—normally involved in maintaining myelin homeostasis—undergo composition alterations, transforming from immunologically inert vesicles into potential triggers of neuroinflammatory responses [94]. Collectively, these findings underscore that EV-mediated crosstalk among glial cells plays a central role in sustaining chronic neuroinflammation in AD.

3.1.3. Dissemination of Oxidative Stress

Oxidative stress represents another key mechanism in AD pathogenesis, intricately linked with Aβ aggregation, tau pathology, and neuroinflammation [95]. Mitochondrial dysfunction is a major source of reactive oxygen species (ROS), functioning as both a cause and a consequence of oxidative damage—a hallmark of neurodegeneration [96,97,98]. Critically, this oxidative burden is not confined to individual cells but is propagated throughout the brain via EVs [99]. Under oxidative stress, neurons release EVs carrying mitochondrial DNA (mtDNA) [100]. Once released into extracellular space, these mtDNA-bearing EVs act as damage-associated molecular patterns (DAMPs) and, upon uptake by recipient microglia or astrocytes, they activate cytoplasmic DNA sensors (primarily the cGAS/STING pathway) and other pattern-recognition receptors (like endosomal TLR9), triggering a robust pro-inflammatory response that further amplifies neuroinflammation [101]. This EV-mediated interplay between oxidative damage and inflammation creates a vicious cycle that synergize with Aβ and tau pathology, ultimately driving synaptic dysfunction and neuronal loss [102]. Compounding this damage, NDEV from AD patients exhibit significantly reduced levels of key functional proteins, these include deficits in synaptic proteins such as neurogranin, synaptophysin, GAP43 [103], along with deficits in stress-response and signaling proteins, collectively impairing neuronal health [38,103,104]. The role of ADEVs in oxidative stress is complex and context-dependent. For instance, in models of amyotrophic lateral sclerosis (ALS), EVs derived from astrocytes expressing mutant SOD1 (e.g., SOD1G93A) transfer misfolded SOD1 to spinal neurons and induce selective motor neuron death, thereby exacerbating oxidative stress and neurodegeneration [64]. This paradox underscores that the functional outcome of EV-mediated transfer is critically dependent on the physiological state and cargo of the parent cell.

3.2. The Bright Side: Endogenous Protective Functions of EVs in AD

In contrast to their role in propagating pathology, EVs also function as vital defense system, preserving neural homeostasis and repair in AD. This section consolidates the multifaceted protective mechanisms that counter the disease processes described earlier, including clearance of toxic proteins, neuroprotection and immunomodulation, preservation of synaptic integrity, and the restoration of metabolic balance. Collectively, these functions position EVs as intrinsic defenders against the progression of AD.

3.2.1. Clearance of Pathogenic Proteins

A primary protective function of EVs is the active clearance of Aβ and the reduction of pathological tau phosphorylation [105,106]. NDEVs can sequester soluble Aβ oligomers, and these EV-bound Aβ complexes are subsequently phagocytosed and degraded by microglia, thereby reducing amyloid plaque burden [107]. This endogenous clearance mechanism can be augmented therapeutically. For instance, statins promote the secretion of exosomes enriched with Insulin-degrading enzyme (IDE). These exosomes then mediate the enhanced degradation of extracellular Aβ, a process demonstrated in microglial cultures and in the circulation of mice [108]. Similarly, a single intracerebral administration of bone marrow-derived MSC-EVs was shown to reduced Aβ plaques in APP/PS1 mice, an effect potentially mediated by the presence of Aβ-degrading enzyme Neprilysin (NEP) in EVs [52]. Furthermore, BMSC-EVs containing GDF-15 upregulated the expression of IDE and NEP, thereby enhancing the clearance of Aβ42 and reduce its intracellular levels [109]. Our own unpublished work using broccoli-derived exosomes (BDEVs) also shows a significant reduction in hippocampus and cortical Aβ levels alongside improved cognitive function in APP/PS1 mice, underscoring the therapeutic potential of EV-mediated clearance [110].
Beyond Aβ, multiple studies have confirmed that EVs offer a potent strategy against tau pathology by targeting its hyperphosphorylation. A key mechanism involves the inhibition of glycogen synthase kinase-3β (GSK-3β) activity, a key kinase driving tau pathology [111,112]. This is evidenced by exosomes from human induced pluripotent stem cells (hiPSC-NSCs) reduced tau phosphorylation in Aβ-exposed neurons by regulating autophagy and GSK-3β activity [113]. Another example is provided by exosome derived from human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), which carry galectin (GAL-3), this cargo suppresses GSK-3β activity, thereby alleviating tau pathology and promote neuronal survival [114]. Collectively, these findings underscore the therapeutic promise of harnessing EVs’ native protective functions against AD.

3.2.2. Neuroprotection and Immunomodulation

EVs exert direct neuroprotective and immunoregulatory effects, primarily by delivering specific cargo that counteracts neurodegeneration and inflammation [115,116]. For instance, MSC-EVs can deliver miRNA, such as miR-29c-3p, which suppresses BACE1 expression and activate the pro-survival Wnt/β-catenin signaling pathway, thereby protecting neurons from Aβ-induced oxidative stress and synaptic damage [117]. In parallel, exosomes from human umbilical cord mesenchymal stem cells (hUC-MSCs-EVs) intrinsically modulate neuroinflammation by shifting the cytokine balance toward an anti-inflammatory state [118,119]. The therapeutic potential of this immunomodulation is further demonstrated by studies showing that intranasal administration of MSC-derived exosomes significantly reduces hippocampus inflammation, enhances neurogenesis in the subventricular zone (SVZ), and lead to cognitive recovery in AD models [120].
Glia-derived EVs play a pivotal role in re-establishing immune homeostasis in the AD brain. When microglia are polarized toward a neuroprotective phenotype (M2) by signals such as interleukin (IL-4), the resulting MDEVs carry TRME2, which activating the Wnt/β-catenin signaling pathway, thereby ameliorating neuronal ferroptosis, oxidative stress and neuroinflammation in AD mouse model [67]. Similarly, ADEVs produced under anti-inflammatory conditions (e.g., IL-10 stimulation) promote neurite outgrowth and neuronal survival [41]. Furthermore, this restorative glia-immune axis can be therapeutically harnessed. For instance, hUC-MSCs-EVs can shift microglia toward an anti-inflammatory state, further enhancing Aβ clearance and reducing neuroinflammation [118]. Together, these findings indicate that endogenous EVs function as physiological messengers of neuroprotection, capable of resetting dysregulated inflammatory networks in AD.

3.2.3. Synaptic Preservation

Synaptic dysfunction and loss is a strong correlate of cognitive decline in AD, and EVs play a direct role in preserving synaptic integrity [121]. In vitro studies using microfluidic chambers have shown that NDEVs are internalized by recipient neurons and can undergo transsynaptic transfer, facilitating inter-neuronal communication and promoting excitatory synaptogenesis [122]. Supporting this concept, exosome derived from neuronal cells or human cerebrospinal fluid can neutralize the synaptotoxic effects of Aβ oligomers, preserving synaptic plasticity and preventing cognitive impairment in AD models [123]. The release and molecular composition of neuronal EVs are closely coupled to neuronal activity. For example, depolarization-induced NDEVs that are enriched with microtubule-associated protein 1b (MAP1b) could further enhance synaptic plasticity and connectivity [124]. Furthermore, glia-derived EVs provide essential support to this neuronal network. ODEVs are critical for maintaining axonal integrity and neuronal health [71]. Notably, ODEVs deliver SIRT2 protein, which has been shown to restore hippocampal neurogenesis and synaptic plasticity through activation of the AKT/GSK-3β signaling pathway [125]. Similarly, MDEVs also contribute to synaptic maintenance in a state-dependent manner, with their release and cargo composition being directly regulated by neuronal activity [126]. This secretion can be influenced by specific signals, including neurotransmitter serotonin (5-HT) and the Wnt3a pathway. Notably, ATP stimulation triggers the secretion of MDEVs enriched with synaptogenic proteins like thrombospondin-1 and thrombospondin-4, which actively promote neurite outgrowth and synapse formation [126,127,128]. Collectively, EVs from neurons and glia form a coordinated communication system that is vital for preserving synaptic integrity in the face of AD pathology.

3.2.4. Alleviation of Oxidative Stress

Oxidative stress, characterized by a persistent imbalance between ROS production and cellular antioxidant capacity, is a critical driver of pathogenesis of AD [129]. Within this context, EVs—particularly those from microglia and oligodendrocytes—have emerged as key regulators of the cerebral redox environment [130,131]. A pivotal mechanism involves the packaging and intercellular delivery of antioxidant enzymes. For instance, Peng et al. demonstrated that MDEVs could mitigate oxidative stress and promote angiogenesis by activating the Keap1/Nrf2/HO-1 signaling pathway, upregulating the antioxidant genes like HO-1, NQO1, Gclc, Cat, and Gsxl [131]. Alongside MDEVs, ODEVs also play an essential role in maintaining redox homeostasis. ODEVs facilitate the intercellular transfer of powerful antioxidant enzymes, including SOD1 and catalase, establishing an essential oligodendrocyte–neuron axis that helps maintain redox balance in the CNS [48]. Furthermore, ADEVs contribute to protection by carrying stress-responsive cargo. This includes heat shock proteins and nucleoside triphosphate diphosphohydrolases (NTPDases), which help mitigate metabolic or thermal stress and support neuronal survival [61,132]. Together, this EV-mediated crosstalk constitutes a fundamental, multi-cellular strategy for the CNS to mitigate oxidative stress and maintain neuronal homeostasis.

3.2.5. Metabolic Modulation and Folate Delivery

Emerging evidence links metabolic disturbances to AD pathogenesis; particularly hyperhomocysteinemia (HHcy), which is widely recognized as a significant risk factor for cognitive decline and AD [133]. A meta-analysis of 35 studies confirmed that blood homocysteine (Hcy) levels in AD patients are approximately one-third higher than in healthy controls [134]. The pathogenic mechanism underlying HHcy in AD involves in the promotion of oxidative stress and neuroinflammation, which directly exacerbate Aβ deposition and tau hyperphosphorylation [135,136,137]. At molecular level, HHcy increases the generation of Hcy-thiolactone (HTL), leading to aberrant protein N-homocysteinylation that disrupts protein structure and induces neurotoxicity [137]. Furthermore, HHcy can drive AD pathology by upregulating the mTOR signaling pathway, thereby inhibiting autophagy and reducing the clearance of Aβ [138]. Notably, HHcy often presents alongside deficiencies in folate and vitamin B12, and this combination exhibits a strong association with AD [139]. This suggests that correcting these nutritional deficiencies could be a viable therapeutic strategy to lower Hcy levels and potentially show disease progression. In addition, epidemiological evidence further support this, indicating that high folate intake is associated with a reduced risk of AD [140]. However, the efficiency of dietary folate is limited by its inefficient delivery to the brain parenchyma.
We hypothesize that the efficient delivery of folate to the brain parenchymal largely depend on the specific recognition of folate-receptor α (FRα), which is highly expressed in MSC-exo, results in efficient delivery of folate reach the neurons or glia. Supporting this hypothesis, recent data in our lab has shown that intranasal administration of low-dose MSC-exo significantly alleviates cognitive deficits and reduce Aβ accumulation in AD mouse model. Crucially, this treatment led to a significant increase in brain 5-Methyltetrahydrofolic acid (5-MTHF) and a concurrent decrease in HCY levels [141]. This preliminary data indicates a promising mechanism whereby MSC-EVs, via their surface FRα, could enhance folate uptake in the brain parenchyma and ultimately counteracting HHcy in AD.
The dual role of EVs in AD is governed by specific contextual factors that determine their function output. (1) Local tissue conditions critically influence EV biogenesis and release: pro-inflammatory signal (TNFα, IL-1β, Aβ) promote the secretion of ADEVs carrying DAMPs, cytokines, and pathogenic proteins, thereby promoting neuroinflammation and oxidative stress [61,91]. Conversely, anti-inflammatory molecules (IL-4, IL-10) or specific neuro-modulatory signals (e.g., 5-HT) favor the release of MDEVs loaded with growth factors, antioxidant enzymes, and miRNA that suppress inflammation and support neuronal repair [126,127]. Metabolic stressors such as HHcy or metabolic dysfunction can also alter EV cargo, lining systemic metabolic disturbances to CNS EV signaling. (2) These extracellular cues ultimately influence parent cell’s identity and physiological state, which directly dictate EV cargo composition and function. For example, MDEVs can be either neurotoxic (when derived from pro-inflammatory M1 microglia) or neuroprotective (from anti-inflammatory M2 phenotype), depending on local cytokines such as damage-associated molecules ATP or IL-4 [67]. Similarly, ADEVs could shift from promoting neurite outgrowth under anti-inflammatory conditions (e.g., IL-10 stimulation) to impairing neuronal function under pro-inflammatory stimuli (e.g., IL-1β or Aβ exposure) [41,126]. (3) Furthermore, the net functional impact of EVs may evolve with AD progression. In the initial stages, EVs may participate in compensatory clearance of Aβ and tau, support synaptic plasticity, and modulate immune surveillance [106,107,117,122]. As pathology progressed, chronic stress and inflammation drive the release of EVs enriched with pathogenic proteins (Aβ, tau) and inflammatory factor, accelerating disease spread and neuronal damage [81,82].
Together, these findings establish EVs as intrinsic guardians of neural homeostasis. Their ability to clear pathogenic proteins, deliver neuroprotective cargo and coordinate intercellular defense mechanisms underpins their promise as naturally evolved therapeutic vehicles.

4. Harnessing EVs for Therapy: Targeting Core AD Pathologies

The complex pathology of AD presents multiple therapeutic targets, with the aggregation of Aβ, hyperphosphorylation of tau, and chronic neuroinflammation being the most prominent. EVs, particularly when engineered, offer innovative and targeted strategies to address these core pathologies. This section explores how EV-based approaches are designed to directly target the key mechanisms driving AD progression.

4.1. Engineered EVs for Targeting Aβ and Tau Pathology

The propagation and accumulation of Aβ and hyperphosphorylated tau are central to AD pathogenesis. EVs are being engineered to enhance the degradation and clearance of these toxic proteins, moving beyond their natural capabilities. For example, to boost Aβ clearance, researchers have developed mannose-modified exosomes loaded with gemfibrozil (MExo-Gem), which specifically target microglia. This strategy enhances their lysosomal activity, leading to reduced amyloid burden and improved cognitive function in AD mice [142]. Beyond Aβ, EVs are also being harnessed to target tau pathology. One study used EVs derived from curcumin-primed macrophages (Exo-cur), which delivered curcumin to the brain and inhibited tau protein hyperphosphorylation via the AKT/GSK-3β pathway [112]. Furthermore, an approach used NDEV engineered to overexpress Fe65, enabling them to deliver the autophagy-inducer Corynoxine-B specifically to APP-expressing neurons. This strategy not only promoted Aβ clearance via autophagy but also ameliorated cognitive deficits by enhancing synaptic regeneration in AD mouse model [143]. These strategies expand upon EV’s natural capacity for Aβ and tau pathology, converting them into programmed nanocarriers for Aβ and tau-targeted therapy.

4.2. Suppressing Neuroinflammation with Engineered EVs

Chronic neuroinflammation, driven by activated microglia and astrocytes, significantly contributes to AD progression. Engineered EVs, particularly those derived from Mesenchymal stem cells (MSC-EVs), have emerged as potent anti-inflammatory delivery systems. For example, human umbilical cord MSC-derived EVs loaded with miR-146a-5p effectively suppress the IRAK1/TRAF6-NF-kB signaling cascade, thereby reducing the production of pro-inflammatory cytokines such as IL-6 and TNFα [144]. Likewise, MSC-derived exosomes engineered with the brain-targeting RVG peptide (MSC-RVG-Exo) attenuate neuroinflammation in AD mouse models by rebalancing the brain’s cytokine profile, suppressing pro-inflammatory factors while promoting anti-inflammatory ones [145]. Moreover, exosomes from MSC preconditioned by hypoxia further ameliorate neuroinflammation by downregulating TNF-α, IL-1β and upregulating of IL-4, IL-10 [146,147]. These immunomodulatory strategies highlight the remarkable versatility of engineered EVs in reprogramming the neuroimmune microenvironment toward a protective, anti-inflammatory state.

4.3. Advanced Engineering: EVs as Delivery Systems for Genetic and Epigenetic Therapy

A next-generation strategy for AD therapy involves using EVs as delivery vehicles for precise genetic interventions, such as the CRISPR/Cas9 systems. This technology holds remarkable potential for targeting genes implicated in AD pathogenesis, such as knocking out MAPT (Tau) to prevent neurofibrillary tangle formation and disrupting GSAP to modulate γ-secretase activity [148]. Moreover, this approach has been used to target AD by directly correcting disease-causing mutations in genes like APP, PSEN1 and PSEN2 to reduce the production of toxic Aβ [148,149,150]. The pioneering work of Alvare-Erviti et al. demonstrated this potential by using engineered exosomes fusing with rabies viral glycoprotein (RVG) peptides to enable specific brain targeting. Injection of these RVG modified exosomes carrying BACE-specific siRNA led to a significant knockdown of BACE1 in the mouse brain [151]. Building on these advances, Han et al. developed a novel photoinducible exosome system (MAPLEX) capable of delivering functional proteins, including a Cas9-DNMT3A epigenome-editing complex targeting BACE1 gene. This approach successfully reduced BACE1 expression, ameliorating memory deficits and decreasing amyloid pathology [152]. Importantly, this study demonstrated that exosomes can safely and efficiently deliver the CRISPR-Cas9 system to target cells while minimizing off-target effects in surrounding tissues [153]. Although CRISPR-Cas9 based EV therapies are still in early developmental stage, continuous optimization of EV loading efficiency and targeting specificity is paving the way for CRISPR-edited EVs to emerge as a next generation precision medicine tool for directly modifying core AD pathologies.

5. EVs as a Diagnostic Tool

Biomarkers are crucial for AD, as neuropathological changes begin years before clinical symptoms appear. Therefore, early diagnosis is crucial for intervention of AD progress. Based on the 2018 National Institute on Aging-Alzheimer’s association (NIA-AA) research framework, defines AD biologically based on three core pathological processes—amyloid beta (A), tau (T) and neurodegeneration (N)—collectively termed the ATN framework [154]. However, this framework currently relies on established cerebrospinal fluid (CSF) analysis and neuroimaging techniques like positron emission tomography (PET) and magnetic resonance imaging (MRI). While informative, these methods are limited by high costs, invasiveness, and limited accessibility, restricting their use for widespread screening and longitudinal monitoring. Consequently, the development of blood-based biomarkers has become a major focus. The plasma Aβ42/40 ratio [155,156,157,158], and phosphorylated tau (p-tau) species, including p-tau181, p-tau217, and p-tau 231 [159,160,161,162,163,164] show promise as A and T biomarkers, respectively, in the AT(N) framework [165,166]. A study identified plasma brain-derived tau (BD-tau) as a novel blood-based biomarker that is strongly correlated with CSF BD-tau, accurately distinguishes autopsy-confirmed AD from other neurodegenerative disease, and associated with Aβ and neurofibrillary tangle [167]. Using the Quanterix Simoa quantification system, a recent Swedish population-based cohort study followed 2148 dementia-free older adults for up to 16 years and found blood level of the biomarkers p-tau181, p-tau217, NFL, and GFP were significantly associated with an increased risk of developing all-cause and Alzheimer’s dementia [168].
Even though blood-based Aβ and p-tau show great potential for diagnosing AD, they present several limitations. The sensitivity of assays measuring blood-based Aβ can be impacted by peripheral sources, and the magnitude of change detected in blood is often less pronounced than in CSF [169,170]. Furthermore, many biomarker-detection methods can be relatively costly for specific patient demographics. Critically, current blood-based biomarkers fail to provide region-specific information about brain pathology, limiting their utility for disease staging and tracking progression [171].
In this context, EVs, particularly those derived from the CNS, have emerged as a revolutionary diagnostic platform. Their unique biological properties—including the ability to cross blood–brain barrier, low immunogenicity, and capacity to carry disease-related biomolecules from their parent cells—make them ideal “liquid biopsies” of the brain [172,173]. The analysis of EV cargo in biofluids like blood or CSF can reveal host cell biological changes, predict dementia onset, reflect cognitive changes, track treatment responses, and evaluate brain recovery [37].

5.1. Neuron-Derived EVs: Capturing Core Pathology and Synaptic Integrity

NDEVs hold significant promises as diagnostic biomarkers because their cargo directly mirrors the physiological and pathological state of neurons. Studies have demonstrated that the levels of pathogenic proteins in NDEVs isolated from peripheral blood strongly correlate with those in CSF and exhibit high diagnostic accuracy for AD [174]. Furthermore, NDEVs levels of Aβ42 increase progressively across the AD continuum—from cognitively normal amyloid-negative individuals to amyloid-positive individuals, to those with mild cognitive impairment (MCI), and finally to AD dementia—and correlate with cerebral amyloid burden on PET imaging, predicting future cognitive decline and entorhinal cortex atrophy [175]. The predictive power of NDEVs is further highlighted by studies showing that a model incorporating p-T181-tau, p-S396-tau, and Aβ1–42 from plasma NDEVs could correctly classified 96.4% of AD patients and predict disease onset up to 10 years before clinical diagnosis [176]. Similarly, abnormal levels of P-tau, Aβ1–42, neurogranin (NRGN), and repressor element 1-silencing transcriptional factor (REST) in plasma NDEVs can accurately predict conversion from MCI to AD dementia [38].
Beyond core pathologies, NDEVs also provide a window into synaptic integrity, the loss of which is a strong correlation of cognitive decline [177]. Synaptic proteins such as NRGN, synaptophysin, synaptopodin within NDEVs are progressively lower in patients with MCI and AD compared to cognitively normal controls (CNC) [38,177]. A more detailed analysis measuring five synaptic proteins in NDEVs (syntaxin 1, GAP43, GluR2, PSD95, and NRGN) revealed a complex signature: while most proteins were reduced, NRGN levels were elevated in early AD patients. A discriminant model incorporating GluR2, proBDNF, NRGN and GAP43 achieved an AD classification accuracy of 81.3%. Crucially, levels of these synaptic proteins correlated significantly with cognitive scores (MMSE and COR-SOB), suggesting they are not only for diagnosis but also for monitoring disease progression [178].

5.2. The Diagnostic Potential of miRNA and Glial Cells-Derived EVs

The diagnostic potential of EVs extends beyond proteins to their nucleic acid cargo. miRNAs within EVs are also potential biomarker candidates due to their stability in extracellular circulation and their involvement in regulating key biological processes in both pathological and physiological state, including neuronal proliferation and development [179,180]. In the AD brain, specific miRNA are dysregulated, including the downregulation of miRNAs that regulate BACE1 and APP (e.g., miR-29a/b-1, miR-107), and the upregulation of those that enhance tau phosphorylation and neuroinflammation (e.g., miR-125b, miR-146a) [181]. Recent reviews have identified 13 upregulated miRNAs and 14 downregulated miRNAs associated with AD pathology [182]. Detecting these dysregulated miRNAs in readily accessible biofluids like plasma and CSF positions EV-associated miRNA as promising, minimally invasive biomarkers for the early diagnosis and tracking of AD.
EV derived from other CNS cell types also offer unique diagnostic insights. The contents of ADEVs vary with disease state, and their associated pathological factors can serve as biomarkers for CNS diseases [84]. For instance, ADEVs from AD patients contain significantly elevated levels of proteins directly involved in Aβ pathology, such as BACE-1 and aAPPβ, which can effectively distinguish AD patients from healthy controls with high accuracy (AUCs of 0.78 and 0.83, respectively) [65]. Furthermore, the complement protein profile within ADEVs undergoes significant alterations during the progression from MCI to AD. Specifically, patients with MCI who converted to AD dementia (MCIC) showed significantly elevated levels of classical and alternative pathway effector proteins—including C1q, C4b, factor D, Bb, C3b, C5b, and the C5b-C9 terminal complement complex—compared to those with stable MCI (MCIS) [183]. These findings not only differentiate AD patients from controls but also exhibit high diagnostic sensitivity for identifying MCI patients at high risk of converting to AD (Table 2).

6. Promising Therapeutic Strategy: Link to Clinical Treatment

The preceding sections have detailed the functional heterogeneity of EVs, underscoring that the role of endogenous EVs in AD is complex and context-dependent. As demonstrated, EVs derived from neurons and glial cells (NDEVs, ADEVs, MDEVs, ODEVs) can paradoxically both propagate pathology and mediate protection, with their function determined by the physiological state of their parent cells. By harnessing EVs from beneficial sources or engineering them to override pathological cues, their innate biological properties can be leveraged for intervention. In contrast to these native vesicles, MSC-EVs have emerged as exogenous therapeutic candidates. They have emerged as particularly promising therapeutic vectors due to their consistently demonstrated protective and regenerative functions in preclinical models. In this section, we outline the key inherent advantages of EVs that position them as next-generation therapeutics for AD.

6.1. Biocompatibility and Low Immunogenicity

EVs exhibit exceptional biocompatibility and a favorable safety profile, primarily due to their endogenous origin, which minimizes immunological reactivity and makes them better tolerated than synthetic nanoparticles [191,192,193,194]. As naturally derived from host cells, exosomes inherently possess low immunogenicity [192]. Studies have shown that MSC-derived EVs maintain low immunogenicity even after repeated administrations, without triggering significant inflammatory responses [195]. Critically, this safety extends to engineered EVs; for instance, systemic administration of exosomes modified with anchor peptides and splicing-switching oligonucleotides result in no detectable toxicity or inflammation in major organs like the liver, kidney, and muscles [196]. This high degree of tolerability is particularly crucial for chronic neurodegenerative diseases like AD, where long-term treatment is required [197]. Furthermore, as non-replicative and non-mutagenic entities, EVs avoids regulatory concerns associated with adverse effects or tumorigenesis [198]. Collectively, their high biocompatibility and established safety profile not only enhances patient tolerance but also provides a strong foundation for the broad application of EVs in regenerative medicine.

6.2. BBB Penetration

The ability of EVs to cross the highly selectively BBB represents a significant therapeutic advantage for treating AD, where the efficient delivery of therapeutic to the brain remains a major obstacle [199]. Owing to their nanoscale size (30–150 nm) and endogenous lipid bilayer membrane, EVs can traverse the BBB through multiple mechanisms, including transcytosis (such as clathrin-mediated, caveola-mediated, and adsorptive-mediated endocytosis), as well as via paracellular pathway when the BBB compromised under pathological conditions [198]. This membrane composition not only protects encapsulated cargo from degradation but also facilitates its stable delivery to brain tissues [200,201]. Preclinical studies have confirmed that intravenously or intranasally administered MSC-EVs accumulate in the brain parenchyma [202,203], where they deliver functional miRNAs and growth factors that alleviate neurodegeneration and promote neurogenesis [203]. Supporting this, our preliminary data indicate that intranasal delivery of MSC-EVs effectively penetrates the BBB and alleviates AD pathology [141]. This intrinsic BBB-penetrating capability confers a clear edge over conventional macromolecules and antibodies, which often fail to reach effective concentrations in the brain, underscoring the inherent advantage of MSC-EVs for treating neurodegenerative disease like AD.

6.3. Regenerative Potential via Wnt/β-Catenin Signaling

Extensive studies have identified dysregulation of Wnt signaling as a contributing factor in AD pathogenesis. For instance, a recent study highlighted that downregulation of Wnt1, PORCN, and RSPO2 in the brains of AD patients inhibits the Wnt/β-catenin signaling pathway [204]. Given its crucial role in AD, Wnt signaling represents a compelling target for therapeutic intervention [205,206,207,208,209]. Beyond functioning as delivery vehicles, MSC-EVs exert regenerative effects by modulating key signaling pathways, particularly Wnt/β-catenin [210]. A key study by Sha et al. provides a direct mechanistic link, demonstrating that BM-MSC-EVs carry and deliver microRNA-29c-3p to neuronal cells. This miRNA subsequently inhibits BACE1, which in turn leads to the activation of the Wnt/β-catenin pathway, ameliorating AD pathology in rat models [117]. This pathway plays a central role in regulating neuronal proliferation, differentiation, survival, and BBB repair, making it a critical mediator of neurogenesis and angiogenesis—processes which are essential for repairing AD-damaged neural circuits [211]. By delivering specific miRNAs and growth factors, MSC-EVs activate the Wnt/β-catenin pathway in recipient cells, leading to the stabilization and nuclear translocation of β-catenin. This, in turn, orchestrates the transcription of genes involved in synaptic plasticity, neuronal survival, and the formation of new blood vessels [212,213]. For instance, adipose-derived MSC exosomes promote angiogenesis in endothelial cells by activating this pathway [214]. Similarly, MSC-EVs support bone regeneration by reducing oxidative stress and restoring cellular proliferation [215], and enhance follicular regeneration through pathway activation [216]. The activation of the Wnt/β-catenin pathway serves as a unifying mechanism, stabilizing β-catenin to coordinate the complex cellular interactions required for effective neural repair. Through enhancing neurogenesis and angiogenesis, MSC-EVs represent an innovative therapeutic strategy for AD by restoring neural integrity and function mediated by Wnt signaling.

6.4. Clinical Translation: Ongoing Trails and Early Outcomes

The therapeutic potential of MSC-EVs, as detailed in the preceding sections, is increasingly being validated in clinical settings. The intrinsic advantages position them as promising candidates for clinical translation, particularly for complex neurodegenerative disorders like AD where treatment options remain limited.
Pioneering clinical trials are now evaluating the safety and efficacy of EV-based therapies across a range of diseases. A notable Phase I//II trial (NCT04388982) is assessing the safety and efficacy of intranasally administered allogeneic adipose-derived MSC-EVs (ahaMSCs-Exos) in patients with mild to moderate AD. Participants received twice-weekly administrations over 12 weeks. Preliminary reports indicate that the treatment was well-tolerated across all dosage groups and was associated with improvements in cognitive function. Notably, the medium-dose group exhibited reduced hippocampal atrophy, suggesting a potential neuroprotective effect. While significant changes in amyloid or tau deposition were not observed across the cohorts, these early outcomes are promising, demonstrating the capacity of MSC-EVs to modulate disease progression and preserve brain structure [217]. Supporting these findings, another clinical study involving 13 AD patients demonstrated that intranasal administration of MSC-EVs over 8 weeks raised no safety concerns and was associated with significant improvement in the Hasegawa Revised Dementia Scale (HDS-R) scores. Increased levels of apolipoprotein, a molecule involved in Aβ clearance, further positioning intranasal MSC-EVs as a safe and convenient therapeutic for AD [218].
Beyond AD-focused studies, other clinical trials provide corroborative evidence for the CNS applicability of EV-based therapies. A Phase I//II clinical trial for acute ischemic stroke (NCT03384433) is evaluating the safety and efficacy of bone marrow MSC-EVs transfected with miR-124. Preliminary data suggests that stereotactic administration is safe, with functional recovery outcomes still under assessment. These promising results highlight the potential of engineered EVs for targeted CNS repair.
The clinical exploration of MSC-EVs extends to other neurological and systemic conditions, reflecting their broad therapeutic utility. Active or completed trials are investigating their use in Parkinson’s disease (NCT05152394), amyotrophic lateral sclerosis (NCT07105371), and psychiatric disorders such as depression and anxiety (NCT04202770). Furthermore, their regenerative and immunomodulatory properties are being harnessed in dermatology for conditions like atopic dermatitis (NCT05969717) and chronic wounds (NCT02565264), as well as in pulmonology for COVID-19-related ARDS, showcasing a versatile and expanding clinical landscape. While larger, multi-center trails with longer follow-up periods are needed to establish their efficacy and optimal dosing regimens, the current clinical data underscore the translational premise of MSC-EVs as a next-generation, cell-free therapeutic strategy for AD.

6.5. Industrial Development and Commercialization of Exosome-Based Therapeutics for AD

In parallel with academic and clinical advances, increasing industrial investment underscores the translational potential of EV-based therapeutics. Several biotechnology companies and small-to-medium enterprises (SMEs), including Aegle Therapeutics [219], Codiak Bioscience [220], and Evox Therapeutics [221] are advancing exosome-based delivery systems designed to enhance tissue targeting, cargo stability, and biological compatibility [222]. In addition, companies such as Exo Biologics have established GMP-compliant manufacturing infrastructure to support the scalable production of EV-based products for clinical and potential commercial needs [223]. Although most commercial programs remain at preclinical or early clinical stages, these initiatives highlight growing confidence in EVs as next-generation delivery systems for neurodegenerative diseases, including AD.

7. Challenges and Limitations on the Path to Translation

Despite the considerable promise of MSC-EV therapy as a cutting-edge “cell-free” regenerative strategy, several major challenges must be addressed before its widespread clinical translation can be realized. These limitations span from production and characterization to delivery and long-term safety.

7.1. Heterogeneity and Standardization

A primary obstacle is the intrinsic heterogeneity of EV populations. Variations in vesicle size, cargo composition (including proteins, lipids, and nucleic acids), and functional properties are influenced by the MSC source, culture conditions, and isolation methods [224]. This biological diversity is further compounded by differences in the physiological and pathological state of the parent cells, resulting in substantial batch-to-batch inconsistency [225,226]. For example, MSCs from the same donor exhibit significant differences in composition, bioactivity, and therapeutic potential among EVs derived from different subtypes [225]. Similarly, EVs derived from other CNS cell types (e.g., astrocytes, neurons, microglia) exhibit divergent or even opposing biological functions, complicating efforts to standardize EV-based interventions [182]. Thus, robust criteria for EV identity, purity, potency and functional characterization remain urgently needed to ensure reproducibility across laboratories and clinical settings.

7.2. Scalable Manufacturing and GMP Compliance

Translating EV therapeutics from bench to bedside demands scalable and reproducible manufacturing pipelines. However, conventional isolation techniques, particularly ultracentrifugation, are often time-consuming, yield inconsistent purity, and are difficult to scale [227,228]. Such inconsistencies in active ingredient content between batches make it difficult to meet the stringent Good Manufacturing Practice (GMP) standards required for drug uniformity [229]. Although alternative methods such as tangential flow filtration offer improved yield, reproducibility, and scalability, further optimization is still needed for robust industrial application [230,231]. Additionally, the large-scale expansion of MSCs under GMP conditions is resource-intensive and costly, significantly limiting the broader availability and commercialization of EV-based therapeutics.

7.3. Targeting Specificity and Delivery Efficiency

Achieving precise targeting of EVs to specific cell types in the complex brain environment is crucial, as it maximizes therapeutic efficacy and minimizes off-target effects. The natural tropism of unmodified EVs is often non-specific, leading to widespread biodistribution and potential dilution of therapeutic impact [232]. Surface engineering approaches, such as decorating EVs membranes with targeting peptides (e.g., RVG peptides for acetylcholine receptors on neuronal cells), have improved uptake in AD-relevant regions in preclinical models [151]. However, the long-term safety, immunogenicity, and functional stability of such engineered EVs in humans still require thorough evaluation. Additionally, the presence of the BBB and the dynamic brain microenvironment further complicates efficient and sustained delivery, underscoring the need to optimize the innovation in EV modification and administration routes.

7.4. Long-Term Safety and Clinical Validation

Although MSC-EVs have demonstrated favorable safety profiles and significant therapeutic efficacy in animal models-such as in AD and mild cognitive impairment (MCI) models, where they effectively improved pathological indicators by modulating immune responses, promoting tissue repair, and suppressing inflammation without observed toxicity. However, their long-term safety and efficacy in human patients remain incompletely established [233]. While clinical trials have shown promising tolerability and preliminary benefits in conditions including COVID-19 [234,235] and Type 1 Diabetes (NCT02138331), most studies are still in their early stages, with limited sample sizes and short follow-up periods. Therefore, to fully validate the therapeutic potential of MSC-EVs, multi-center, large-scale, and long-term clinical trials are necessary to determine optimal dosing regimens (e.g., dose, frequency, and administration route) and establish standardized preparation protocols to ensure the quality of EVs. Concurrently, fundamental research should be strengthened to elucidate their mechanisms of action and interactions with MSC-EVs, providing a scientific basis for their safety and effective translation from the laboratory to the clinic.

8. Future Perspectives

Looking ahead, several emerging technologies and integrated strategies hold significant potential to advance the use of EVs in diagnosis and treatment of AD. These approaches aim to overcome existing limitations in specificity, delivery, and therapeutic efficacy, paving the way for more precise and powerful interventions (Figure 2).

8.1. Microfluidic Chip for Blood-Based EVs Diagnostics

Microfluidic technology represents a promising strategy for developing blood-based diagnostic platforms for AD. Since 2010, microfluidic systems have been fabricated for exosome manipulation, offering unique advantages such as small sample volume, low cost, product purity, and short operation time [236]. For instance, a ciliated micropillar microfluidic device was designed to traps exosome-sized vesicles via size-selective filtration [237]. In one notable application, an integrated microfluidic chip was developed that combines immunoisolation with on-chip analysis of plasma-derived exosomes, establishing a promising liquid biopsy platform for non-invasive cancer diagnosis [238]. Building on this, the same research group created an ExoSearch chip for the immunomagnetic isolation and multiplexed detection of circulating exosomes, enabling blood-based diagnosis of ovarian cancer through simultaneous quantification of three exosomal tumor markers [239].
Looking forward, this lab-on-chip technology could be strategically adapted to address critical challenges in AD diagnostics. By utilizing antibodies against neuron-specific surface markers (e.g., L1CAM), such a chip could selectively enrich NDEVs from peripheral blood, serving as a real-time “liquid biopsy” of the brain. Furthermore, the integrated design would allow for the subsequent on-chip lysis of captured NDEVs and the quantitative analysis of intravascular protein targets relevant to AD pathogenesis (e.g., Aβ, p-tau, p-tau217). This microfluidic approach could thus act as a powerful and practical tool for the early detection of molecular changes in AD, potentially identifying at-risk individuals’ years before clinical onset.

8.2. Focused Ultrasound to Enhance EV Therapy

Focused ultrasound (FUS) has emerged as a promising non-invasive and highly controllable technique to transiently open the BBB. This capability provides a powerful means to enhance drug delivery to target brain regions in AD. Preclinical and clinical studies have demonstrated that MRI-guided FUS could safely, reversibly, and repeatedly open the BBB in AD animal model even in human patients, without inducing significant tissue damage [240,241]. Complementing these findings, our recent work using a BBB-on-a-chip microfluidic platform provided a quantitative and physiological relevant system to assess FUS-induced BBB modulation. This platform confirmed that FUS markedly enhanced trans-BBB transport, serving as a robust tool for evaluating FUS-based therapeutic strategies [242].
Beyond its role in facilitating drug delivery, FUS exerts several intrinsic therapeutic effects relevant to AD pathology. For instance, scanning ultrasound alone has been shown to remove Aβ and restore cognitive function in AD mouse model [243]. Additional studies have demonstrated that FUS mitigates AD-related pathology and improves spatial memory in mouse models and patients [244]. A recent clinical study further supports the therapeutic potential of FUS, revealing that combinatory therapy using FUS and aducanumab (anti-amyloid antibody) significantly reduces Aβ aggregation in sonicated brain regions of AD patients [245].
Mechanistic investigations suggest that these therapeutic benefits may be partly mediated through glia activation: microglia and astrocytes in FUS-treated cortical regions display increases protein expression and morphological changes indicative of activation [246]. FUS also influences intercellular communication. Deng et al. reported that ultrasound stimulation of astrocytes led to a 5-fold increase in EVs release. These ultrasound-primed ADEVs were rapidly taken up by neurons, resulting in reduced internalization of Aβ42 and thus decreasing its neurotoxicity in vitro [247]. Moreover, FUS has been shown to enhance glymphatic transport—a glia-dependent perivascular clearance pathway—which may further facilitate the removal of pathological proteins such as Aβ [248].
Collectively, these findings position FUS as a promising modality for AD therapy, functioning both as an enabling technology for enhanced drug delivery and as a biologically active intervention that directly modulates pathology. The continued integration of FUS with EVs creates a powerful synergistic strategy: FUS provides the non-invasive, spatially precise key to unlock the BBB, while EVs serve as ideal natural nanocarriers, leveraging their innate biocompatibility and efficient BBB-penetrating ability to deliver therapeutics. This combination of precise physical access and sophisticated biological delivery is poised to significantly amplify therapeutic impact and accelerate the translation of these technologies into clinical practice.

9. Conclusions

EVs are increasingly recognized as critical dual agents in AD, serving as both propagators of pathology and revolutionary platforms for diagnosis and therapy. While their endogenous role in disseminating Aβ, tau, and inflammatory signals exacerbates disease, their innate biocompatibility, ability to cross the BBB, and capacity to deliver bioactive cargo highlight their potential as next-generation interventions. Therapeutically, MSC-derived EVs and engineered EVs have demonstrated the ability to promote clearance of toxicological protein, suppressing chronic neuroinflammation, and delivering targeted genetic or pharmacological cargo directly to the brain. In parallel, EV-based “liquid biopsies” are transforming the diagnostic landscape, providing minimally invasive biomarkers—such as EV-associated proteins and miRNAs—for early detection, tracking disease progression, and monitoring of therapeutic responses. Despite these advances, key challenges—including EV heterogeneity, scalable GMP manufacturing, targeted delivery efficiency, and long-term safety—must be resolved before clinical translation can be fully realized. Future directions will advance early detection platforms via EV-based microfluidic systems and pioneer the integration of EVs with FUS. This combinatorial approach will enable spatiotemporally controlled EVs delivery and amplify therapeutic efficacy. Encouragingly, early-phase clinical studies report favorable safety profiles and initial cognitive improvements, underscoring the feasibility of EV-based approaches.
In summary, EVs represent a rapidly advancing and clinically relevant frontier in AD research. As standardization and precision engineering continue to mature, EV-based diagnostics and therapeutics are poised to become integral components of future AD management, offering meaningful opportunities to modify disease courses and improve patient outcomes.

Author Contributions

Conceptualization: Y.L. and H.F.; writing—original draft: F.L. and Y.L.; writing—reviewing and editing: H.F., Y.L., F.L., L.W. and X.F.; Software: L.W.; Validation: X.F., Y.L. and H.F.; Visualization: L.W., Y.L. and H.F.; Supervision: Y.L. and H.F.; Funding acquisition: Y.L. and H.F. Resources: Y.L. and H.F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Zhejiang Provincial Natural Science Foundation of China (LQ24H160003), the Medical Scientific Research Foundation of Zhejiang Province (2023KY299 and 2024KY351), the Ningbo Natural Science Foundation (2023J365), the Young Innovative Talent Project of Yongjiang Talent Introduction Programme of Ningbo Municipal Government (2021A-012-G), and the Bei’An Talent Programme of Jiangbei District of Ningbo (2023RC004).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADAlzheimer’s disease
amyloid-β
APPamyloid precursor protein
ADEVsAstrocyte-derived extracellular vesicles
ALSamyotrophic lateral sclerosis
ATNamyloid beta (A), tau (T) and neurodegeneration (N)
ahaMSCs-Exosadministered allogeneic adipose-derived MSC-EVs
BD-taubrain-derived tau
BDENsbroccoli-derived exosomes-like nanoparticles
BACE-1β-secretase
CNScentral nervous system
CNP2′,3′-cyclic nucleotide 3′-phosphodiesterase
CSFcerebrospinal fluid
CNCcognitively normal controls
DAMPsdamage-associated molecular patterns
Exo-curEVs derived from curcumin-primed macrophages
EVsextracellular vesicles
EAAT1excitatory amino acid transporter 1
FRαfolate-receptor α
FUSfocused ultrasound
GFAPglial fibrillary acidic protein
GSK-3βglycogen synthase kinase-3β
GMPgood manufacturing practice
GAL-3galectin
hUCB-MSCshuman umbilical cord blood-derived mesenchymal stem cells
hUC-MSCs-EVsexosomes from human umbilical cord mesenchymal stem cells
hiPSC-NSCshuman induced pluripotent stem cells
HHcyhyperhomocysteinemia
Hcyhomocysteine
HTLhcy-thiolactone
HDS-RHasegawa revised dementia scale
IDEinsulin-degrading enzyme
ILVsintraluminal vesicles
IL-4interleukin-4
L1CAML1 cell adhesion molecule
MSCmesenchymal stem cell
MVBsmultivesicular bodies
MDEVsmicroglia-derived extracellular vesicles
MHCmajor histocompatibility complex
MBPmyelin basic protein
MSC-EVsmesenchymal stem cell-derived extracellular vesicles
mtDNAmitochondrial DNA
MOGmyelin oligodendrocyte glycoprotein
MSCsmesenchymal stem cells
MAP1bmicrotubule-associated protein 1b
MExo-Gemmannose-modified exosomes loaded with gemfibrozil
MSC-RVG-ExoMSC-derived exosomes engineered with the brain-targeting RVG peptide
MAPLEXa novel photoinducible exosome system
MRImagnetic resonance imaging
miRNAsmicroRNAs
MCImild cognitive impairment
MCICpatients with MCI who converted to AD dementia
MCISstable MCI
NDEVsneuron-derived extracellular vesicles
NEPneprilysin
NIA-AANational Institute on aging-Alzheimer’s association
NRGNneurogranin
NFTsneurofibrillary tangles
NTPDasesnucleoside triphosphate diphosphohydrolases
ODEVsoligodendrocyte-derived extracellular vesicles
PLPproteolipid protein
p-tauphosphorylated tau
PETpositron emission tomography
ROSreactive oxygen species
RVGrabies viral glycoprotein
RESTrepressor element 1-silencing transcriptional
sAPPβsoluble amyloid precursor protein β
SVZsubventricular zone
SCDsubjective cognitive decline
VaDvascular dementia
5-MTHF5-Methyltetrahydrofolic acid
5-HTserotonin

References

  1. Better, M.A. 2024 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2024, 20, 3708–3821. [Google Scholar]
  2. Malaha, K.A.; Thebaut, C.; Achille, D.; Preux, P.M.; Guerchet, M. Costs of Dementia in Low- and Middle-Income Countries: A Systematic Review. J. Alzheimer’s Dis. 2023, 91, 115–128. [Google Scholar] [CrossRef]
  3. Rayaprolu, S.; Higginbotham, L.; Bagchi, P.; Watson, C.M.; Zhang, T.; Levey, A.I.; Rangaraju, S.; Seyfried, N.T. Systems-based proteomics to resolve the biology of Alzheimer’s disease beyond amyloid and tau. Neuropsychopharmacology 2020, 46, 98–115. [Google Scholar] [CrossRef]
  4. Abdelnour, C.; Agosta, F.; Bozzali, M.; Fougere, B.; Iwata, A.; Nilforooshan, R.; Takada, L.T.; Vinuela, F.; Traber, M. Perspectives and challenges in patient stratification in Alzheimer’s disease. Alzheimer’s Res. Ther. 2022, 14, 112. [Google Scholar] [CrossRef]
  5. Bloom, G.S. Amyloid-beta and tau: The trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 2014, 71, 505–508. [Google Scholar] [CrossRef]
  6. Zhang, J.F.; Zhang, Y.L.; Wang, J.X.; Xia, Y.L.; Zhang, J.X.; Chen, L. Recent advances in Alzheimer’s disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct. Target. Ther. 2024, 9, 211. [Google Scholar] [CrossRef] [PubMed]
  7. Zheng, Q.; Wang, X. Alzheimer’s disease: Insights into pathology, molecular mechanisms, and therapy. Protein Cell 2025, 16, 83–120. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, Y.; Chen, H.; Li, R.; Sterling, K.; Song, W. Amyloid beta-based therapy for Alzheimer’s disease: Challenges, successes and future. Signal Transduct. Target. Ther. 2023, 8, 248. [Google Scholar] [CrossRef]
  9. Twarowski, B.; Herbet, M. Inflammatory Processes in Alzheimer’s Disease—Pathomechanism, Diagnosis and Treatment: A Review. Int. J. Mol. Sci. 2023, 24, 6518. [Google Scholar] [CrossRef] [PubMed]
  10. Heneka, M.T.; van der Flier, W.M.; Jessen, F.; Hoozemanns, J.; Thal, D.R.; Boche, D.; Brosseron, F.; Teunissen, C.; Zetterberg, H.; Jacobs, A.H.; et al. Neuroinflammation in Alzheimer disease. Nat. Rev. Immunol. 2024, 25, 321–352. [Google Scholar] [CrossRef]
  11. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2020, 17, 157–172. [Google Scholar] [CrossRef]
  12. Klegeris, A.; Bajwa, E. Neuroinflammation as a mechanism linking hypertension with the increased risk of Alzheimer’s disease. Neural Regen. Res. 2022, 17, 2342–2346. [Google Scholar] [CrossRef] [PubMed]
  13. Busche, M.A.; Hyman, B.T. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat. Neurosci. 2020, 23, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, H.; Wei, W.; Zhao, M.; Ma, L.; Jiang, X.; Pei, H.; Cao, Y.; Li, H. Interaction between Aβ and Tau in the Pathogenesis of Alzheimer’s Disease. Int. J. Biol. Sci. 2021, 17, 2181–2192. [Google Scholar] [CrossRef]
  15. Kumar, M.A.; Baba, S.K.; Sadida, H.Q.; Marzooqi, S.A.; Jerobin, J.; Altemani, F.H.; Algehainy, N.; Alanazi, M.A.; Abou-Samra, A.-B.; Kumar, R.; et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct. Target. Ther. 2024, 9, 27. [Google Scholar] [CrossRef]
  16. Belkozhayev, A.M.; Al-Yozbaki, M.; George, A.; Ye Niyazova, R.; Sharipov, K.O.; Byrne, L.J.; Wilson, C.M. Extracellular Vesicles, Stem Cells and the Role of miRNAs in Neurodegeneration. Curr. Neuropharmacol. 2022, 20, 1450–1478. [Google Scholar] [CrossRef]
  17. 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]
  18. Marar, C.; Starich, B.; Wirtz, D. Extracellular vesicles in immunomodulation and tumor progression. Nat. Immunol. 2021, 22, 560–570. [Google Scholar] [CrossRef]
  19. Buratta, S.; Tancini, B.; Sagini, K.; Delo, F.; Chiaradia, E.; Urbanelli, L.; Emiliani, C. Lysosomal Exocytosis, Exosome Release and Secretory Autophagy: The Autophagic- and Endo-Lysosomal Systems Go Extracellular. Int. J. Mol. Sci. 2020, 21, 2576. [Google Scholar] [CrossRef]
  20. Lee, Y.; Andaloussi, S.E.; Wood, M.J. Exosomes and microvesicles: Extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef]
  21. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e18. [Google Scholar] [CrossRef]
  22. Wang, X.; Tian, L.; Lu, J.G.; Ng, I.O.L. Exosomes and cancer—Diagnostic and prognostic biomarkers and therapeutic vehicle. Oncogenesis 2022, 11, 54. [Google Scholar] [CrossRef] [PubMed]
  23. Dreyer, F.; Baur, A. Biogenesis and Functions of Exosomes and Extracellular Vesicles. Methods Mol. Biol. 2016, 1448, 201–216. [Google Scholar]
  24. Ratajczak, M.Z.; Ratajczak, J. Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia 2020, 34, 3126–3135. [Google Scholar] [CrossRef]
  25. Poon, I.K.H.; Parkes, M.A.F.; Jiang, L.; Atkin-Smith, G.K.; Tixeira, R.; Gregory, C.D.; Ozkocak, D.C.; Rutter, S.F.; Caruso, S.; Santavanond, J.P.; et al. Moving beyond size and phosphatidylserine exposure: Evidence for a diversity of apoptotic cell-derived extracellular vesicles in vitro. J. Extracell Vesicles 2019, 8, 1608786. [Google Scholar] [CrossRef]
  26. Santavanond, J.P.; Rutter, S.F.; Atkin-Smith, G.K.; Poon, I.K.H. Apoptotic Bodies: Mechanism of Formation, Isolation and Functional Relevance. Subcell Biochem. 2021, 97, 61–88. [Google Scholar]
  27. Estevez-Souto, V.; Da Silva-Alvarez, S.; Collado, M. The role of extracellular vesicles in cellular senescence. FEBS J. 2023, 290, 1203–1211. [Google Scholar] [CrossRef]
  28. Dixson, A.; Dawson, T.R.; Di Vizio, D.; Weaver, A.M. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat. Rev. Mol. Cell Biol. 2023, 24, 454–476. [Google Scholar] [CrossRef] [PubMed]
  29. Joshi, P.; Benussi, L.; Furlan, R.; Ghidoni, R.; Verderio, C. Extracellular vesicles in Alzheimer’s disease: Friends or foes? Focus on abeta-vesicle interaction. Int. J. Mol. Sci. 2015, 16, 4800–4813. [Google Scholar] [CrossRef] [PubMed]
  30. Li, Z.; Wang, X.; Wang, X.; Yi, X.; Wong, Y.K.; Wu, J.; Xie, F.; Hu, D.; Wang, Q.; Wang, J.; et al. Research progress on the role of extracellular vesicles in neurodegenerative diseases. Transl. Neurodegener. 2023, 12, 43. [Google Scholar] [CrossRef]
  31. Kalluri, R. The biology and function of extracellular vesicles in immune response and immunity. Immunity 2024, 57, 1752–1768. [Google Scholar] [CrossRef]
  32. Chen, J.; Tian, C.; Xiong, X.; Yang, Y.; Zhang, J. Extracellular vesicles: New horizons in neurodegeneration. eBioMedicine 2025, 113, 105605. [Google Scholar] [CrossRef]
  33. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  34. Mathivanan, S.; Ji, H.; Simpson, R.J. Exosomes: Extracellular organelles important in intercellular communication. J. Proteomics 2010, 73, 1907–1920. [Google Scholar] [CrossRef] [PubMed]
  35. Antonucci, F.; Turola, E.; Riganti, L.; Caleo, M.; Gabrielli, M.; Perrotta, C.; Novellino, L.; Clementi, E.; Giussani, P.; Viani, P.; et al. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J. 2012, 31, 1231–1240. [Google Scholar] [CrossRef] [PubMed]
  36. Manolopoulos, A.; Delgado-Peraza, F.; Mustapic, M.; Pucha, K.A.; Nogueras-Ortiz, C.; Daskalopoulos, A.; Knight, D.D.; Leoutsakos, J.M.; Oh, E.S.; Lyketsos, C.G.; et al. Comparative assessment of Alzheimer’s disease-related biomarkers in plasma and neuron-derived extracellular vesicles: A nested case-control study. Front. Mol. Biosci. 2023, 10, 1254834. [Google Scholar] [CrossRef]
  37. Gonul, C.P.; Karacicek, B.; Genc, S. Neuron-Derived Extracellular Vesicles: Emerging Biomarkers and Functional Mediators in Alzheimer’s Disease, with Comparative Insights into Neurodevelopment and Aging. Dev. Neurobiol. 2025, 85, e22984. [Google Scholar] [CrossRef]
  38. Winston, C.N.; Goetzl, E.J.; Akers, J.C.; Carter, B.S.; Rockenstein, E.M.; Galasko, D.; Masliah, E.; Rissman, R.A. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimer’s Dement. 2016, 3, 63–72. [Google Scholar] [CrossRef]
  39. Gonzalez-Molina, L.A.; Villar-Vesga, J.; Henao-Restrepo, J.; Villegas, A.; Lopera, F.; Cardona-Gomez, G.P.; Posada-Duque, R. Extracellular Vesicles From 3xTg-AD Mouse and Alzheimer’s Disease Patient Astrocytes Impair Neuroglial and Vascular Components. Front. Aging Neurosci. 2021, 13, 593927. [Google Scholar] [CrossRef]
  40. Kawata, K.; Mitsuhashi, M.; Aldret, R. A Preliminary Report on Brain-Derived Extracellular Vesicle as Novel Blood Biomarkers for Sport-Related Concussions. Front. Neurol. 2018, 9, 239. [Google Scholar] [CrossRef]
  41. Li, B.; Ma, Z.; Li, Z. A novel regulator in Alzheimer’s disease progression: The astrocyte-derived extracellular vesicles. Ageing Res. Rev. 2023, 86, 101871. [Google Scholar] [CrossRef]
  42. Zhang, Y.; Xu, C.; Nan, Y.; Nan, S. Microglia-Derived Extracellular Vesicles Carrying miR-711 Alleviate Neurodegeneration in a Murine Alzheimer’s Disease Model by Binding to Itpkb. Front. Cell Dev. Biol. 2020, 8, 566530. [Google Scholar] [CrossRef]
  43. Potolicchio, I.; Carven, G.J.; Xu, X.; Stipp, C.; Riese, R.J.; Stern, L.J.; Santambrogio, L. Proteomic analysis of microglia-derived exosomes: Metabolic role of the aminopeptidase CD13 in neuropeptide catabolism. J. Immunol. 2005, 175, 2237–2243. [Google Scholar] [CrossRef]
  44. Hanisch, U.K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387–1394. [Google Scholar] [CrossRef]
  45. Kramer-Albers, E.M.; Bretz, N.; Tenzer, S.; Winterstein, C.; Mobius, W.; Berger, H.; Nave, K.A.; Schild, H.; Trotter, J. Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: Trophic support for axons? PROTEOMICS Clin. Appl. 2007, 1, 1446–1461. [Google Scholar] [CrossRef]
  46. Bakhti, M.; Winter, C.; Simons, M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J. Biol. Chem. 2011, 286, 787–796. [Google Scholar] [CrossRef] [PubMed]
  47. Kramer-Albers, E.M.; Werner, H.B. Mechanisms of axonal support by oligodendrocyte-derived extracellular vesicles. Nat. Rev. Neurosci. 2023, 24, 474–486. [Google Scholar] [CrossRef] [PubMed]
  48. Frohlich, D.; Kuo, W.P.; Fruhbeis, C.; Sun, J.J.; Zehendner, C.M.; Luhmann, H.J.; Pinto, S.; Toedling, J.; Trotter, J.; Kramer-Albers, E.M. Multifaceted effects of oligodendroglial exosomes on neurons: Impact on neuronal firing rate, signal transduction and gene regulation. Philos. Trans. R Soc. Lond. B Biol. Sci. 2014, 369, 20130510. [Google Scholar] [CrossRef] [PubMed]
  49. Bodart-Santos, V.; de Carvalho, L.R.P.; de Godoy, M.A.; Batista, A.F.; Saraiva, L.M.; Lima, L.G.; Abreu, C.A.; De Felice, F.G.; Galina, A.; Mendez-Otero, R.; et al. Extracellular vesicles derived from human Wharton’s jelly mesenchymal stem cells protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-beta oligomers. Stem. Cell Res. Ther. 2019, 10, 332. [Google Scholar] [CrossRef]
  50. Gorabi, A.M.; Kiaie, N.; Barreto, G.E.; Read, M.I.; Tafti, H.A.; Sahebkar, A. The Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes in Treatment of Neurodegenerative Diseases. Mol. Neurobiol. 2019, 56, 8157–8167. [Google Scholar] [CrossRef]
  51. Zhang, X.; Che, X.; Zhang, S.; Wang, R.; Li, M.; Jin, Y.; Wang, T.; Song, Y. Mesenchymal stem cell-derived extracellular vesicles for human diseases. Extracell Vesicles Circ. Nucl. Acids 2024, 5, 64–82. [Google Scholar] [CrossRef] [PubMed]
  52. 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 Abeta Plaque Burden in Early Stages of a Preclinical Model of Alzheimer’s Disease. Cells 2019, 8, 1059. [Google Scholar] [CrossRef]
  53. Bravo-Miana, R.D.C.; Arizaga-Echebarria, J.K.; Otaegui, D. Central nervous system-derived extracellular vesicles: The next generation of neural circulating biomarkers? Transl. Neurodegener. 2024, 13, 32. [Google Scholar] [CrossRef]
  54. Amin, S.; Massoumi, H.; Tewari, D.; Roy, A.; Chaudhuri, M.; Jazayerli, C.; Krishan, A.; Singh, M.; Soleimani, M.; Karaca, E.E.; et al. Cell Type-Specific Extracellular Vesicles and Their Impact on Health and Disease. Int. J. Mol. Sci. 2024, 25, 2730. [Google Scholar] [CrossRef]
  55. Lemaire, Q.; Duhamel, M.; Raffo-Romero, A.; Salzet, M.; Lefebvre, C. Characterization of Immune Cell-derived Extracellular Vesicles and Studying Functional Impact on Cell Environment. J. Vis. Exp. 2020, 160, e60118. [Google Scholar] [CrossRef]
  56. Jana, K.; Ghosh, S.; Parua, P.; Debnath, B.; Halder, J.; Sahoo, R.K.; Rai, V.K.; Pradhan, D.; Dash, P.; Das, C.; et al. Insights into the Versatile Role of Extracellular Vesicles in the Treatment of CNS Disorders. Mol. Neurobiol. 2025, 63, 14. [Google Scholar] [CrossRef] [PubMed]
  57. Chinnathambi, S.; Kumarappan, M.; Malik, S.; Velmurugan, G.; Chandrashekar, M. Neuron-derived extracellular vesicle-based diagnostics for Tau and Alzheimer’s disease. Brain Netw. Disord. 2025, 1, 150–166. [Google Scholar] [CrossRef]
  58. Huo, L.; Du, X.; Li, X.; Liu, S.; Xu, Y. The Emerging Role of Neural Cell-Derived Exosomes in Intercellular Communication in Health and Neurodegenerative Diseases. Front. Neurosci. 2021, 15, 738442. [Google Scholar] [CrossRef]
  59. Solana-Balaguer, J.; Campoy-Campos, G.; Martin-Flores, N.; Perez-Sisques, L.; Sitja-Roqueta, L.; Kucukerden, M.; Gamez-Valero, A.; Coll-Manzano, A.; Marti, E.; Perez-Navarro, E.; et al. Neuron-derived extracellular vesicles contain synaptic proteins, promote spine formation, activate TrkB-mediated signalling and preserve neuronal complexity. J. Extracell Vesicles 2023, 12, e12355. [Google Scholar] [CrossRef] [PubMed]
  60. Wang, X.; Li, A.; Fan, H.; Li, Y.; Yang, N.; Tang, Y. Astrocyte-Derived Extracellular Vesicles for Ischemic Stroke: Therapeutic Potential and Prospective. Aging Dis. 2024, 15, 1227–1254. [Google Scholar]
  61. Chaudhuri, A.D.; Dasgheyb, R.M.; DeVine, L.R.; Bi, H.; Cole, R.N.; Haughey, N.J. Stimulus-dependent modifications in astrocyte-derived extracellular vesicle cargo regulate neuronal excitability. Glia 2020, 68, 128–144. [Google Scholar] [CrossRef]
  62. Li, J.; Xu, H.; Zhang, K.; Liu, Y.; Zeng, C.; Fu, Y.; Li, Y. Astrocyte-derived exosomes-transported miRNA-26a-5p ameliorates sevoflurane-induced cognitive dysfunction in aged mice. Transl. Res. 2024, 268, 79–96. [Google Scholar] [CrossRef]
  63. Long, X.; Yao, X.; Jiang, Q.; Yang, Y.; He, X.; Tian, W.; Zhao, K.; Zhang, H. Astrocyte-derived exosomes enriched with miR-873a-5p inhibit neuroinflammation via microglia phenotype modulation after traumatic brain injury. J. Neuroinflamm. 2020, 17, 89. [Google Scholar] [CrossRef]
  64. Basso, M.; Pozzi, S.; Tortarolo, M.; Fiordaliso, F.; Bisighini, C.; Pasetto, L.; Spaltro, G.; Lidonnici, D.; Gensano, F.; Battaglia, E.; et al. Mutant copper-zinc superoxide dismutase (SOD1) induces protein secretion pathway alterations and exosome release in astrocytes: Implications for disease spreading and motor neuron pathology in amyotrophic lateral sclerosis. J. Biol. Chem. 2013, 288, 15699–15711. [Google Scholar] [CrossRef] [PubMed]
  65. Goetzl, E.J.; Mustapic, M.; Kapogiannis, D.; Eitan, E.; Lobach, I.V.; Goetzl, L.; Schwartz, J.B.; Miller, B.L. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J. 2016, 30, 3853–3859. [Google Scholar] [CrossRef]
  66. Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Thery, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef]
  67. Zhu, L.; Zhou, T.; Wu, L.; Zhu, X.; Chen, L.; Zhang, M.; Zhou, J.; Wang, F. Microglial exosome TREM2 ameliorates ferroptosis and neuroinflammation in alzheimer’s disease by activating the Wnt/beta-catenin signaling. Sci. Rep. 2025, 15, 24968. [Google Scholar] [CrossRef] [PubMed]
  68. Guo, M.; Wang, J.; Zhao, Y.; Feng, Y.; Han, S.; Dong, Q.; Cui, M.; Tieu, K. Microglial exosomes facilitate alpha-synuclein transmission in Parkinson’s disease. Brain 2020, 143, 1476–1497. [Google Scholar] [CrossRef]
  69. Wu, J.; Lu, J.; Pan, M.Z.; Gu, X.C.; Dai, L.; Wang, Y.; Shen, B.; Zhang, X.B. Update on the roles and applications of extracellular vesicles in depression. World J. Psychiatry 2025, 15, 102643. [Google Scholar] [CrossRef]
  70. Fruhbeis, C.; Frohlich, D.; Kuo, W.P.; Amphornrat, J.; Thilemann, S.; Saab, A.S.; Kirchhoff, F.; Mobius, W.; Goebbels, S.; Nave, K.A.; et al. Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 2013, 11, e1001604. [Google Scholar] [CrossRef] [PubMed]
  71. Fruhbeis, C.; Kuo-Elsner, W.P.; Muller, C.; Barth, K.; Peris, L.; Tenzer, S.; Mobius, W.; Werner, H.B.; Nave, K.A.; Frohlich, D.; et al. Oligodendrocytes support axonal transport and maintenance via exosome secretion. PLoS Biol. 2020, 18, e3000621. [Google Scholar] [CrossRef] [PubMed]
  72. Simons, M.; Nave, K.A. Oligodendrocytes: Myelination and Axonal Support. Cold Spring Harb. Perspect. Biol. 2015, 8, a020479. [Google Scholar] [CrossRef]
  73. Padinharayil, H.; Varghese, J.; Wilson, C.; George, A. Mesenchymal stem cell-derived exosomes: Characteristics and applications in disease pathology and management. Life Sci. 2024, 342, 122542. [Google Scholar] [CrossRef]
  74. Lu, C.H.; Chen, Y.A.; Ke, C.C.; Liu, R.S. Mesenchymal Stem Cell-Derived Extracellular Vesicle: A Promising Alternative Therapy for Osteoporosis. Int. J. Mol. Sci. 2021, 22, 12750. [Google Scholar] [CrossRef]
  75. Ahn, S.Y.; Park, W.S.; Kim, Y.E.; Sung, D.K.; Sung, S.I.; Ahn, J.Y.; Chang, Y.S. Vascular endothelial growth factor mediates the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicles against neonatal hyperoxic lung injury. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef]
  76. Kou, M.; Huang, L.; Yang, J.; Chiang, Z.; Chen, S.; Liu, J.; Guo, L.; Zhang, X.; Zhou, X.; Xu, X.; et al. Mesenchymal stem cell-derived extracellular vesicles for immunomodulation and regeneration: A next generation therapeutic tool? Cell Death Dis. 2022, 13, 580. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, Y.; Zhang, X.; Zhang, H.; Song, P.; Pan, W.; Xu, P.; Wang, G.; Hu, P.; Wang, Z.; Huang, K.; et al. Mesenchymal Stem Cells Derived Extracellular Vesicles Alleviate Traumatic Hemorrhagic Shock Induced Hepatic Injury via IL-10/PTPN22-Mediated M2 Kupffer Cell Polarization. Front. Immunol. 2021, 12, 811164. [Google Scholar] [CrossRef]
  78. Racchetti, G.; Meldolesi, J. Extracellular Vesicles of Mesenchymal Stem Cells: Therapeutic Properties Discovered with Extraordinary Success. Biomedicines 2021, 9, 667. [Google Scholar] [CrossRef]
  79. Qiu, G.; Zheng, G.; Ge, M.; Wang, J.; Huang, R.; Shu, Q.; Xu, J. Mesenchymal stem cell-derived extracellular vesicles affect disease outcomes via transfer of microRNAs. Stem. Cell Res. Ther. 2018, 9, 320. [Google Scholar] [CrossRef]
  80. Lai, R.C.; Tan, S.S.; Yeo, R.W.; Choo, A.B.; Reiner, A.T.; Su, Y.; Shen, Y.; Fu, Z.; Alexander, L.; Sze, S.K.; et al. MSC secretes at least 3 EV types each with a unique permutation of membrane lipid, protein and RNA. J. Extracell Vesicles 2016, 5, 29828. [Google Scholar] [CrossRef] [PubMed]
  81. Watson, L.S.; Hamlett, E.D.; Stone, T.D.; Sims-Robinson, C. Neuronally derived extracellular vesicles: An emerging tool for understanding Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 22. [Google Scholar] [CrossRef]
  82. Upadhya, R.; Zingg, W.; Shetty, S.; Shetty, A.K. Astrocyte-derived extracellular vesicles: Neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J. Control Release 2020, 323, 225–239. [Google Scholar] [CrossRef]
  83. Wang, L.; Lin, Y.; Yang, Z.; Zhang, K.; Gong, H.; Zheng, Y.; Wang, B.; Zhang, X.; Sun, M. Microglia-derived extracellular vesicles mediate fine particulate matter-induced Alzheimer’s disease-like behaviors through the miR-34a-5p/DUSP10/p-p38 MAPK pathway. J. Hazard Mater. 2025, 495, 138853. [Google Scholar] [CrossRef]
  84. Zhao, S.; Sheng, S.; Wang, Y.; Ding, L.; Xu, X.; Xia, X.; Zheng, J.C. Astrocyte-derived extracellular vesicles: A double-edged sword in central nervous system disorders. Neurosci. Biobehav. Rev. 2021, 125, 148–159. [Google Scholar] [CrossRef] [PubMed]
  85. Guo, M.; Hao, Y.; Feng, Y.; Li, H.; Mao, Y.; Dong, Q.; Cui, M. Microglial Exosomes in Neurodegenerative Disease. Front. Mol. Neurosci. 2021, 14, 630808. [Google Scholar] [CrossRef]
  86. Sasmita, A.O.; Depp, C.; Nazarenko, T.; Sun, T.; Siems, S.B.; Ong, E.C.; Nkeh, Y.B.; Bohler, C.; Yu, X.; Bues, B.; et al. Oligodendrocytes produce amyloid-beta and contribute to plaque formation alongside neurons in Alzheimer’s disease model mice. Nat. Neurosci. 2024, 27, 1668–1674. [Google Scholar] [CrossRef]
  87. Rajani, R.M.; Ellingford, R.; Hellmuth, M.; Harris, S.S.; Taso, O.S.; Graykowski, D.; Lam, F.K.W.; Arber, C.; Fertan, E.; Danial, J.S.H.; et al. Selective suppression of oligodendrocyte-derived amyloid beta rescues neuronal dysfunction in Alzheimer’s disease. PLoS Biol. 2024, 22, e3002727. [Google Scholar] [CrossRef] [PubMed]
  88. Gupta, A.; Pulliam, L. Exosomes as mediators of neuroinflammation. J. Neuroinflamm. 2014, 11, 68. [Google Scholar] [CrossRef]
  89. Lombardi, M.; Gabrielli, M.; Adinolfi, E.; Verderio, C. Role of ATP in Extracellular Vesicle Biogenesis and Dynamics. Front. Pharmacol. 2021, 12, 654023. [Google Scholar] [CrossRef] [PubMed]
  90. Bianco, F.; Pravettoni, E.; Colombo, A.; Schenk, U.; Möller, T.; Matteoli, M.; Verderio, C. Astrocyte-Derived ATP Induces Vesicle Shedding and IL-1 β Release from Microglia. J. Immunol. 2005, 174, 7268–7277. [Google Scholar] [CrossRef]
  91. Wang, G.; Dinkins, M.; He, Q.; Zhu, G.; Poirier, C.; Campbell, A.; Mayer-Proschel, M.; Bieberich, E. Astrocytes secrete exosomes enriched with proapoptotic ceramide and prostate apoptosis response 4 (PAR-4): Potential mechanism of apoptosis induction in Alzheimer disease (AD). J. Biol. Chem. 2012, 287, 21384–21395. [Google Scholar] [CrossRef] [PubMed]
  92. Rajendran, L.; Bali, J.; Barr, M.M.; Court, F.A.; Kramer-Albers, E.M.; Picou, F.; Raposo, G.; van der Vos, K.E.; van Niel, G.; Wang, J.; et al. Emerging roles of extracellular vesicles in the nervous system. J. Neurosci. 2014, 34, 15482–15489. [Google Scholar] [CrossRef]
  93. Dickens, A.M.; Tovar, Y.R.L.B.; Yoo, S.W.; Trout, A.L.; Bae, M.; Kanmogne, M.; Megra, B.; Williams, D.W.; Witwer, K.W.; Gacias, M.; et al. Astrocyte-shed extracellular vesicles regulate the peripheral leukocyte response to inflammatory brain lesions. Sci. Signal 2017, 10, eaai7696. [Google Scholar] [CrossRef]
  94. Yu, Z.; Shi, M.; Stewart, T.; Fernagut, P.O.; Huang, Y.; Tian, C.; Dehay, B.; Atik, A.; Yang, D.; De Giorgi, F.; et al. Reduced oligodendrocyte exosome secretion in multiple system atrophy involves SNARE dysfunction. Brain 2020, 143, 1780–1797. [Google Scholar] [CrossRef]
  95. Butterfield, D.A.; Halliwell, B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat. Rev. Neurosci. 2019, 20, 148–160. [Google Scholar] [CrossRef]
  96. Swerdlow, R.H. Mitochondria and Mitochondrial Cascades in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1403–1416. [Google Scholar] [CrossRef]
  97. Passos, J.F.; Saretzki, G.; Ahmed, S.; Nelson, G.; Richter, T.; Peters, H.; Wappler, I.; Birket, M.J.; Harold, G.; Schaeuble, K.; et al. Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol. 2007, 5, e110. [Google Scholar] [CrossRef] [PubMed]
  98. Gibson, G.E.; Starkov, A.; Blass, J.P.; Ratan, R.R.; Beal, M.F. Cause and consequence: Mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim. Biophys. Acta 2010, 1802, 122–134. [Google Scholar] [CrossRef]
  99. Bir, A.; Ghosh, A.; Chauhan, A.; Saha, S.; Saini, A.K.; Bisaglia, M.; Chakrabarti, S. Exosomal Dynamics and Brain Redox Imbalance: Implications in Alzheimer’s Disease Pathology and Diagnosis. Antioxidants 2024, 13, 316. [Google Scholar] [CrossRef] [PubMed]
  100. Zhao, Y.; Liu, B.; Xu, L.; Yu, S.; Fu, J.; Wang, J.; Yan, X.; Su, J. ROS-Induced mtDNA Release: The Emerging Messenger for Communication between Neurons and Innate Immune Cells during Neurodegenerative Disorder Progression. Antioxidants 2021, 10, 1917. [Google Scholar] [CrossRef] [PubMed]
  101. Liu, Y.; Zhang, B.; Duan, R.; Liu, Y. Mitochondrial DNA Leakage and cGas/STING Pathway in Microglia: Crosstalk Between Neuroinflammation and Neurodegeneration. Neuroscience 2024, 548, 1–8. [Google Scholar] [CrossRef]
  102. Brites, D.; Fernandes, A. Neuroinflammation and Depression: Microglia Activation, Extracellular Microvesicles and microRNA Dysregulation. Front. Cell Neurosci. 2015, 9, 476. [Google Scholar] [CrossRef] [PubMed]
  103. Goetzl, E.J.; Kapogiannis, D.; Schwartz, J.B.; Lobach, I.V.; Goetzl, L.; Abner, E.L.; Jicha, G.A.; Karydas, A.M.; Boxer, A.; Miller, B.L. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016, 30, 4141–4148. [Google Scholar] [CrossRef] [PubMed]
  104. Goetzl, E.J.; Boxer, A.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Miller, B.L.; Carlson, O.D.; Mustapic, M.; Kapogiannis, D. Low neural exosomal levels of cellular survival factors in Alzheimer’s disease. Ann. Clin. Transl. Neurol. 2015, 2, 769–773. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, G.L.; Yang, Y.M.; Zhang, X.Y.; Shi, Z.Q.; Gao, H.L.; Zhong, M.L.; Fan, Y.G.; Zhang, H.Y.; Liu, B.; Qing, G.Y. Secreted endogenous macrosomes reduce Aβ burden and ameliorate Alzheimer’s disease. Sci. Adv. 2023, 21, eade0293. [Google Scholar] [CrossRef]
  106. Zhang, X.; Tang, L.; Yang, J.; Meng, L.; Chen, J.; Zhou, L.; Wang, J.; Xiong, M.; Zhang, Z. Soluble TREM2 ameliorates tau phosphorylation and cognitive deficits through activating transgelin-2 in Alzheimer’s disease. Nat. Commun. 2023, 14, 6670. [Google Scholar] [CrossRef]
  107. Yuyama, K.; Igarashi, Y. Exosomes as Carriers of Alzheimer’s Amyloid-ss. Front. Neurosci. 2017, 11, 229. [Google Scholar] [CrossRef]
  108. Tamboli, I.Y.; Barth, E.; Christian, L.; Siepmann, M.; Kumar, S.; Singh, S.; Tolksdorf, K.; Heneka, M.T.; Lutjohann, D.; Wunderlich, P.; et al. Statins promote the degradation of extracellular amyloid beta-peptide by microglia via stimulation of exosome-associated insulin-degrading enzyme (IDE) secretion. J. Biol. Chem. 2010, 285, 37405–37414. [Google Scholar] [CrossRef]
  109. Xiong, W.P.; Yao, W.Q.; Wang, B.; Liu, K. BMSCs-exosomes containing GDF-15 alleviated SH-SY5Y cell injury model of Alzheimer’s disease via AKT/GSK-3beta/beta-catenin. Brain Res. Bull. 2021, 177, 92–102. [Google Scholar] [CrossRef]
  110. Feng, X.; Fan, H. Ningbo Institute of Life and Health Industry, UCAS, Ningbo, Zhejiang Province, China. 2026; manuscript in preparation. [Google Scholar]
  111. Santos, R.T.; Braga, C.L.; de Sa Freire Onofre, M.E.; da Silva, C.M.; de Novaes Rocha, N.; Veras, R.G.; de Souza Serra, S.S.; Teixeira, D.E.; Dos Santos Alves, S.A.; Miranda, B.T.; et al. Cardioprotective effects of extracellular vesicles from hypoxia-preconditioned mesenchymal stromal cells in experimental pulmonary arterial hypertension. Stem Cell Res. Ther. 2025, 16, 466. [Google Scholar] [CrossRef]
  112. Wang, H.; Sui, H.; Zheng, Y.; Jiang, Y.; Shi, Y.; Liang, J.; Zhao, L. Curcumin-primed exosomes potently ameliorate cognitive function in AD mice by inhibiting hyperphosphorylation of the Tau protein through the AKT/GSK-3beta pathway. Nanoscale 2019, 11, 7481–7496. [Google Scholar] [CrossRef]
  113. Rao, S.; Madhu, L.N.; Babu, R.S.; Shankar, G.; Kotian, S.; Nagarajan, A.; Upadhya, R.; Narvekar, E.; Cai, J.J.; Shetty, A.K. Extracellular vesicles from hiPSC-derived NSCs protect human neurons against Abeta-42 oligomers induced neurodegeneration, mitochondrial dysfunction and tau phosphorylation. Stem Cell Res. Ther. 2025, 16, 191. [Google Scholar] [CrossRef]
  114. Lim, H.; Lee, D.; Choi, W.K.; Choi, S.J.; Oh, W.; Kim, D.H. Galectin-3 Secreted by Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Reduces Aberrant Tau Phosphorylation in an Alzheimer Disease Model. Stem Cells Int. 2020, 2020, 8878412. [Google Scholar] [CrossRef] [PubMed]
  115. de Godoy, M.A.; Saraiva, L.M.; de Carvalho, L.R.P.; Vasconcelos-Dos-Santos, A.; Beiral, H.J.V.; Ramos, A.B.; Silva, L.R.P.; Leal, R.B.; Monteiro, V.H.S.; Braga, C.V.; et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-beta oligomers. J. Biol. Chem. 2018, 293, 1957–1975. [Google Scholar] [CrossRef] [PubMed]
  116. Ma, X.; Huang, M.; Zheng, M.; Dai, C.; Song, Q.; Zhang, Q.; Li, Q.; Gu, X.; Chen, H.; Jiang, G.; et al. ADSCs-derived extracellular vesicles alleviate neuronal damage, promote neurogenesis and rescue memory loss in mice with Alzheimer’s disease. J. Control Release 2020, 327, 688–702. [Google Scholar] [CrossRef] [PubMed]
  117. Sha, S.; Shen, X.L.; Cao, Y.P.; Qu, L. Mesenchymal stem cells-derived extracellular vesicles ameliorate Alzheimer’s disease in rat models via the microRNA-29c-3p/BACE1 axis and the Wnt/β-catenin pathway. Aging 2021, 13, 15285–15306. [Google Scholar] [CrossRef]
  118. 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]
  119. Long, Q.; Upadhya, D.; Hattiangady, B.; Kim, D.K.; An, S.Y.; Shuai, B.; Prockop, D.J.; Shetty, A.K. Intranasal MSC-derived A1-exosomes ease inflammation, and prevent abnormal neurogenesis and memory dysfunction after status epilepticus. Proc. Natl. Acad. Sci. USA 2017, 114, E3536–E3545. [Google Scholar] [CrossRef]
  120. Reza-Zaldivar, E.E.; Hernandez-Sapiens, M.A.; Gutierrez-Mercado, Y.K.; Sandoval-Avila, S.; Gomez-Pinedo, U.; Marquez-Aguirre, A.L.; Vazquez-Mendez, E.; Padilla-Camberos, E.; Canales-Aguirre, A.A. 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]
  121. Tzioras, M.; McGeachan, R.I.; Durrant, C.S.; Spires-Jones, T.L. Synaptic degeneration in Alzheimer disease. Nat. Rev. Neurol. 2022, 19, 19–38. [Google Scholar] [CrossRef]
  122. Antoniou, A.; Auderset, L.; Kaurani, L.; Sebastian, E.; Zeng, Y.; Allahham, M.; Cases-Cunillera, S.; Schoch, S.; Grundemann, J.; Fischer, A.; et al. Neuronal extracellular vesicles and associated microRNAs induce circuit connectivity downstream BDNF. Cell Rep. 2023, 42, 112063. [Google Scholar] [CrossRef] [PubMed]
  123. An, K.; Klyubin, L.; Kim, Y.; Jung, J.H.; Mably, J.A.; O’Dowd, Y.S.; Lynch, T.; Kanmert, D.; Lemere, A.C.; Finan, M.G.; et al. Exosomes neutralize synaptic-plasticity-disrupting activity of Aβ assemblies in vivo. Mol. Brain 2013, 6, 47. [Google Scholar] [CrossRef]
  124. Goldie, B.J.; Dun, M.D.; Lin, M.; Smith, N.D.; Verrills, N.M.; Dayas, C.V.; Cairns, M.J. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Res. 2014, 42, 9195–9208. [Google Scholar] [CrossRef]
  125. Zhang, H.; Xie, X.H.; Xu, S.X.; Wang, C.; Sun, S.; Song, X.; Li, R.; Li, N.; Feng, Y.; Duan, H.; et al. Oligodendrocyte-derived exosomes-containing SIRT2 ameliorates depressive-like behaviors and restores hippocampal neurogenesis and synaptic plasticity via the AKT/GSK-3beta pathway in depressed mice. CNS Neurosci. Ther. 2024, 30, e14661. [Google Scholar] [CrossRef]
  126. Mancini, W.V.S.B.; Mattera, V.S.; Pasquini, J.M.; Pasquini, L.A.; Correale, J.D. Microglia-derived extracellular vesicles in homeostasis and demyelination/remyelination processes. J. Neurochem. 2024, 168, 3–25. [Google Scholar]
  127. Glebov, K.; Lochner, M.; Jabs, R.; Lau, T.; Merkel, O.; Schloss, P.; Steinhauser, C.; Walter, J. Serotonin stimulates secretion of exosomes from microglia cells. Glia 2015, 63, 626–634. [Google Scholar] [CrossRef]
  128. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 polarization and metabolic states. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef]
  129. Bai, R.; Guo, J.; Ye, X.Y.; Xie, Y.; Xie, T. Oxidative stress: The core pathogenesis and mechanism of Alzheimer’s disease. Ageing Res. Rev. 2022, 77, 101619. [Google Scholar] [CrossRef]
  130. Li, S.; Sheng, Z.H. Oligodendrocyte-derived transcellular signaling regulates axonal energy metabolism. Curr. Opin. Neurobiol. 2023, 80, 102722. [Google Scholar] [CrossRef] [PubMed]
  131. Peng, W.; Wan, L.; Luo, Z.; Xie, Y.; Liu, Y.; Huang, T.; Lu, H.; Hu, J.; Xiong, Y. Microglia-Derived Exosomes Improve Spinal Cord Functional Recovery after Injury via Inhibiting Oxidative Stress and Promoting the Survival and Function of Endothelia Cells. Oxid. Med. Cell. Longev. 2021, 2021, 1695087. [Google Scholar] [CrossRef] [PubMed]
  132. Ceruti, S.; Colombo, L.; Magni, G.; Vigano, F.; Boccazzi, M.; Deli, M.A.; Sperlagh, B.; Abbracchio, M.P.; Kittel, A. Oxygen-glucose deprivation increases the enzymatic activity and the microvesicle-mediated release of ectonucleotidases in the cells composing the blood-brain barrier. Neurochem. Int. 2011, 59, 259–271. [Google Scholar] [CrossRef]
  133. Zeng, P.; Shi, Y.; Wang, X.M.; Lin, L.; Du, Y.J.; Tang, N.; Wang, Q.; Fang, Y.Y.; Wang, J.Z.; Zhou, X.W.; et al. Emodin Rescued Hyperhomocysteinemia-Induced Dementia and Alzheimer’s Disease-Like Features in Rats. Int. J. Neuropsychopharmacol. 2019, 22, 57–70. [Google Scholar] [CrossRef] [PubMed]
  134. Zhang, L.; Xie, X.; Sun, Y.; Zhou, F. Blood and CSF Homocysteine Levels in Alzheimer’s Disease: A Meta-Analysis and Meta-Regression of Case-Control Studies. Neuropsychiatr. Dis. Treat. 2022, 18, 2391–2403. [Google Scholar] [CrossRef] [PubMed]
  135. Tawfik, A.; Samra, Y.A.; Elsherbiny, N.M.; Al-Shabrawey, M. Implication of Hyperhomocysteinemia in Blood Retinal Barrier (BRB) Dysfunction. Biomolecules 2020, 10, 1119. [Google Scholar] [CrossRef]
  136. Persichilli, S.; Gervasoni, J.; Di Napoli, A.; Fuso, A.; Nicolia, V.; Giardina, B.; Scarpa, S.; Desiderio, C.; Cavallaro, R.A. Plasma thiols levels in Alzheimer’s disease mice under diet-induced hyperhomocysteinemia: Effect of S-adenosylmethionine and superoxide-dismutase supplementation. J. Alzheimer’s Dis. 2015, 44, 1323–1331. [Google Scholar] [CrossRef]
  137. Jakubowski, H. Homocysteine Thiolactone Detoxifying Enzymes and Alzheimer’s Disease. Int. J. Mol. Sci. 2024, 25, 8095. [Google Scholar] [CrossRef]
  138. Witucki, L.; Borowczyk, K.; Suszynska-Zajczyk, J.; Warzych, E.; Pawlak, P.; Jakubowski, H. Deletion of the Homocysteine Thiolactone Detoxifying Enzyme Bleomycin Hydrolase, in Mice, Causes Memory and Neurological Deficits and Worsens Alzheimer’s Disease-Related Behavioral and Biochemical Traits in the 5xFAD Model of Alzheimer’s Disease. J. Alzheimer’s Dis. 2023, 95, 1735–1755. [Google Scholar] [CrossRef]
  139. Chen, H.; Liu, S.; Ji, L.; Wu, T.; Ma, F.; Ji, Y.; Zhou, Y.; Zheng, M.; Zhang, M.; Huang, G. Associations between Alzheimer’s Disease and Blood Homocysteine, Vitamin B12, and Folate: A Case-Control Study. Curr. Alzheimer Res. 2015, 12, 88–94. [Google Scholar] [CrossRef]
  140. Gu, Y.; Nieves, W.J.; Stern, Y.; Luchsinger, A.P.; Scarmeas, N. Food Combination and Alzheimer Disease Risk. Arch. Neurol. 2010, 7, 699–706. [Google Scholar] [CrossRef]
  141. Li, F.; Li, Y. Ningbo Institute of Life and Health Industry, UCAS, Ningbo, Zhejiang Province, China. 2026; manuscript in preparation. [Google Scholar]
  142. Hao, Y.; Su, C.; Liu, X.; Sui, H.; Shi, Y.; Zhao, L. Bioengineered microglia-targeted exosomes facilitate Abeta clearance via enhancing activity of microglial lysosome for promoting cognitive recovery in Alzheimer’s disease. Biomater. Adv. 2022, 136, 212770. [Google Scholar] [CrossRef] [PubMed]
  143. Iyaswamy, A.; Thakur, A.; Guan, X.J.; Krishnamoorthi, S.; Fung, T.Y.; Lu, K.; Gaurav, I.; Yang, Z.; Su, C.F.; Lau, K.F.; et al. Fe65-engineered neuronal exosomes encapsulating corynoxine-B ameliorate cognition and pathology of Alzheimer’s disease. Signal Transduct. Target. Ther. 2023, 8, 404. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, J.; Xu, Z.; Yu, J.; Zang, X.; Jiang, S.; Xu, S.; Wang, W.; Hong, S. MiR-146a-5p engineered hucMSC-derived extracellular vesicles attenuate Dermatophagoides farinae-induced allergic airway epithelial cell inflammation. Front. Immunol. 2024, 15, 1443166. [Google Scholar] [CrossRef] [PubMed]
  145. Cui, G.H.; Guo, H.D.; Li, H.; Zhai, Y.; Gong, Z.B.; Wu, J.; Liu, J.S.; Dong, Y.R.; Hou, S.X.; Liu, J.R. RVG-modified exosomes derived from mesenchymal stem cells rescue memory deficits by regulating inflammatory responses in a mouse model of Alzheimer’s disease. Immun. Ageing 2019, 16, 10. [Google Scholar] [CrossRef]
  146. Cui, G.H.; Wu, J.; Mou, F.F.; Xie, W.H.; Wang, F.B.; Wang, Q.L.; Fang, J.; Xu, Y.W.; Dong, Y.R.; Liu, J.R.; et al. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J. 2018, 32, 654–668. [Google Scholar] [CrossRef] [PubMed]
  147. Ni, H.; Yang, S.; Siaw-Debrah, F.; Hu, J.; Wu, K.; He, Z.; Yang, J.; Pan, S.; Lin, X.; Ye, H.; et al. Exosomes Derived From Bone Mesenchymal Stem Cells Ameliorate Early Inflammatory Responses Following Traumatic Brain Injury. Front. Neurosci. 2019, 13, 14. [Google Scholar] [CrossRef]
  148. Bhardwaj, S.; Kesari, K.K.; Rachamalla, M.; Mani, S.; Ashraf, G.M.; Jha, S.K.; Kumar, P.; Ambasta, R.K.; Dureja, H.; Devkota, H.P.; et al. CRISPR/Cas9 gene editing: New hope for Alzheimer’s disease therapeutics. J. Adv. Res. 2022, 40, 207–221. [Google Scholar] [CrossRef]
  149. Akyuz, E.; Aslan, F.S.; Gokce, E.; Ilmaz, O.; Topcu, F.; Kakac, S. Extracellular vesicle and CRISPR gene therapy: Current applications in Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, and Huntington’s disease. Eur. J. Neurosci. 2024, 60, 6057–6090. [Google Scholar] [CrossRef]
  150. Hanafy, A.S.; Schoch, S.; Lamprecht, A. CRISPR/Cas9 Delivery Potentials in Alzheimer’s Disease Management: A Mini Review. Pharmaceutics 2020, 12, 801. [Google Scholar] [CrossRef]
  151. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
  152. Han, J.; Sul, H.J.; Lee, J.; Kim, E.; Kim, K.H.; Chae, M.; Lim, J.; Kim, J.; Kim, C.; Kim, J.K.; et al. Engineered exosomes with a photoinducible protein delivery system enable CRISPR-Cas–based epigenome editing in Alzheimer’s disease. Drug Deliv. 2024, 16, eadi4830. [Google Scholar] [CrossRef]
  153. Kostyushev, D.; Kostyusheva, A.; Brezgin, S.; Smirnov, V.; Volchkova, E.; Lukashev, A.; Chulanov, V. Gene Editing by Extracellular Vesicles. Int. J. Mol. Sci. 2020, 21, 7362. [Google Scholar] [CrossRef]
  154. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. Contributors, NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  155. Keshavan, A.; Pannee, J.; Karikari, T.K.; Rodriguez, J.L.; Ashton, N.J.; Nicholas, J.M.; Cash, D.M.; Coath, W.; Lane, C.A.; Parker, T.D.; et al. Population-based blood screening for preclinical Alzheimer’s disease in a British birth cohort at age 70. Brain 2021, 144, 434–449. [Google Scholar] [PubMed]
  156. Janelidze, S.; Teunissen, C.E.; Zetterberg, H.; Allue, J.A.; Sarasa, L.; Eichenlaub, U.; Bittner, T.; Ovod, V.; Verberk, I.M.W.; Toba, K.; et al. Head-to-Head Comparison of 8 Plasma Amyloid-beta 42/40 Assays in Alzheimer Disease. JAMA Neurol. 2021, 78, 1375–1382. [Google Scholar] [CrossRef]
  157. Nakamura, A.; Kaneko, N.; Villemagne, V.L.; Kato, T.; Doecke, J.; Dore, V.; Fowler, C.; Li, Q.X.; Martins, R.; Rowe, C.; et al. High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature 2018, 554, 249–254. [Google Scholar] [CrossRef]
  158. Schindler, S.E.; Bollinger, J.G.; Ovod, V.; Mawuenyega, K.G.; Li, Y.; Gordon, B.A.; Holtzman, D.M.; Morris, J.C.; Benzinger, T.L.S.; Xiong, C.; et al. High-precision plasma beta-amyloid 42/40 predicts current and future brain amyloidosis. Neurology 2019, 93, e1647–e1659. [Google Scholar] [CrossRef] [PubMed]
  159. Karikari, T.K.; Pascoal, T.A.; Ashton, N.J.; Janelidze, S.; Benedet, A.L.; Rodriguez, J.L.; Chamoun, M.; Savard, M.; Kang, M.S.; Therriault, J.; et al. Blood phosphorylated tau 181 as a biomarker for Alzheimer’s disease: A diagnostic performance and prediction modelling study using data from four prospective cohorts. Lancet Neurol. 2020, 19, 422–433. [Google Scholar] [CrossRef]
  160. Palmqvist, S.; Janelidze, S.; Quiroz, Y.T.; Zetterberg, H.; Lopera, F.; Stomrud, E.; Su, Y.; Chen, Y.; Serrano, G.E.; Leuzy, A.; et al. Discriminative Accuracy of Plasma Phospho-tau217 for Alzheimer Disease vs Other Neurodegenerative Disorders. JAMA 2020, 324, 772–781. [Google Scholar] [CrossRef]
  161. Janelidze, S.; Mattsson, N.; Palmqvist, S.; Smith, R.; Beach, T.G.; Serrano, G.E.; Chai, X.; Proctor, N.K.; Eichenlaub, U.; Zetterberg, H.; et al. Plasma P-tau181 in Alzheimer’s disease: Relationship to other biomarkers, differential diagnosis, neuropathology and longitudinal progression to Alzheimer’s dementia. Nat. Med. 2020, 26, 379–386. [Google Scholar] [CrossRef] [PubMed]
  162. Thijssen, E.H.; La Joie, R.; Wolf, A.; Strom, A.; Wang, P.; Iaccarino, L.; Bourakova, V.; Cobigo, Y.; Heuer, H.; Spina, S.; et al. Treatment for Frontotemporal Lobar Degeneration, i., Diagnostic value of plasma phosphorylated tau181 in Alzheimer’s disease and frontotemporal lobar degeneration. Nat. Med. 2020, 26, 387–397. [Google Scholar] [CrossRef]
  163. Jiao, B.; Ouyang, Z.; Xiao, X.; Zhang, C.; Xu, T.; Yang, Q.; Zhu, Y.; Liu, Y.; Liu, X.; Zhou, Y.; et al. Development and validation of machine learning models with blood-based digital biomarkers for Alzheimer’s disease diagnosis: A multicohort diagnostic study. eClinicalMedicine 2025, 81, 103142. [Google Scholar] [CrossRef]
  164. Ashton, N.J.; Pascoal, T.A.; Karikari, T.K.; Benedet, A.L.; Lantero-Rodriguez, J.; Brinkmalm, G.; Snellman, A.; Scholl, M.; Troakes, C.; Hye, A.; et al. Plasma p-tau231: A new biomarker for incipient Alzheimer’s disease pathology. Acta Neuropathol. 2021, 141, 709–724. [Google Scholar] [CrossRef]
  165. Karikari, T.K.; Ashton, N.J.; Brinkmalm, G.; Brum, W.S.; Benedet, A.L.; Montoliu-Gaya, L.; Lantero-Rodriguez, J.; Pascoal, T.A.; Suarez-Calvet, M.; Rosa-Neto, P.; et al. Blood phospho-tau in Alzheimer disease: Analysis, interpretation, and clinical utility. Nat. Rev. Neurol. 2022, 18, 400–418. [Google Scholar] [CrossRef]
  166. Ashton, N.J.; Leuzy, A.; Karikari, T.K.; Mattsson-Carlgren, N.; Dodich, A.; Boccardi, M.; Corre, J.; Drzezga, A.; Nordberg, A.; Ossenkoppele, R.; et al. The validation status of blood biomarkers of amyloid and phospho-tau assessed with the 5-phase development framework for AD biomarkers. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 2140–2156. [Google Scholar] [CrossRef]
  167. Gonzalez-Ortiz, F.; Kirsebom, B.E.; Contador, J.; Tanley, J.E.; Selnes, P.; Gisladottir, B.; Palhaugen, L.; Suhr Hemminghyth, M.; Jarholm, J.; Skogseth, R.; et al. Plasma brain-derived tau is an amyloid-associated neurodegeneration biomarker in Alzheimer’s disease. Nat. Commun. 2024, 15, 2908. [Google Scholar] [CrossRef] [PubMed]
  168. Grande, G.; Qiu, C.; Valletta, M.; Orsini4, N.; Rizzuto, D.; Dale, M.; Fredolini, C.; Winblad, B.; Vetrano, D.L. Blood-based biomarkers of Alzheimer’s disease and incident dementia in the community. Nat. Med. 2025, 31, 2027–2035. [Google Scholar] [CrossRef]
  169. Olsson, B.; Lautner, R.; Andreasson, U.; Öhrfelt, A.; Portelius, E.; Bjerke, M.; Hölttä, M.; Rosén, C.; Olsson, C.; Strobel, G.; et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: A systematic review and meta-analysis. Lancet Neurol. 2016, 15, 673–684. [Google Scholar] [CrossRef]
  170. Cleary, A.J.; Kumar, A.; Craft, S. Neuron-derived extracellular vesicles as a liquid biopsy for brain insulin dysregulation in Alzheimer’s disease and related disorders. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2024, 21, e14497. [Google Scholar] [CrossRef]
  171. Zetterberg, H.; Blennow, K. Moving fluid biomarkers for Alzheimer’s disease from research tools to routine clinical diagnostics. Mol. Neurodegener. 2021, 16, 10. [Google Scholar] [CrossRef] [PubMed]
  172. Hagey, D.W.; El Andaloussi, S. The promise and challenges of extracellular vesicles in the diagnosis of neurodegenerative diseases. Handb. Clin. Neurol. 2023, 193, 227–241. [Google Scholar] [PubMed]
  173. Kandeel, M.; Morsy, M.A.; Alkhodair, K.M.; Alhojaily, S. Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities. Biomolecules 2023, 13, 1250. [Google Scholar] [CrossRef]
  174. Jia, L.; Qiu, Q.; Zhang, H.; Chu, L.; Du, Y.; Zhang, J.; Zhou, C.; Liang, F.; Shi, S.; Wang, S.; et al. Concordance between the assessment of Ab42, T-tau, and P-T181-tau in peripheral blood neuronal-derived exosomes and cerebrospinal fluid. Alzheimer’s Dement. 2019, 15, 1071–1080. [Google Scholar] [CrossRef]
  175. Li, T.R.; Yao, Y.; Jiang, X.; Dong, Q.; Yu, W.; Wang, T.; Cai, Y.; Han, Y. β-Amyloid in blood neuronal-derived extracellular vesicles is elevated in cognitively normal adults at risk of Alzheimer’s disease and predicts cerebral amyloidosis. Alzheimer’s Res. Ther. 2022, 14, 66. [Google Scholar] [CrossRef] [PubMed]
  176. Fiandacaa, M.S.; Kapogiannisb, D.; Mapstonec, M.; Boxerd, A.; Eitanb, E.; Schwartze, J.B.; Abnerf, E.L.; Peterseng, R.C.; Federoffa, H.J.; Millerd, B.L.; et al. Identification of pre-clinical Alzheimer’s disease by a profile of pathogenic proteins in neurally-derived blood exosomes: A case-control study. Alzheimer’s Dement. 2014, 11, 600–607. [Google Scholar] [CrossRef]
  177. Winston, C.N.; Goetzl, E.J.; Baker, L.D.; Vitiello, M.V.; Rissman, R.A. Growth Hormone-Releasing Hormone Modulation of Neuronal Exosome Biomarkers in Mild Cognitive Impairment. J. Alzheimer’s Dis. 2018, 66, 971–981. [Google Scholar] [CrossRef]
  178. Eitan, E.; Thornton-Wells, T.; Elgart, K.; Erden, E.; Gershun, E.; Levine, A.; Volpert, O.; Azadeh, M.; Smith, D.G.; Kapogiannis, D. Synaptic proteins in neuron-derived extracellular vesicles as biomarkers for Alzheimer’s disease: Novel methodology and clinical proof of concept. Extracell. Vesicles Circ. Nucleic Acids 2023, 4, 133–150. [Google Scholar] [CrossRef] [PubMed]
  179. Zhang, H.; Yang, H.; Zhang, C.; Jing, Y.; Wang, C.; Liu, C.; Zhang, R.; Wang, J.; Zhang, J.; Zen, K.; et al. Investigation of microRNA expression in human serum during the aging process. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2015, 70, 102–109. [Google Scholar] [CrossRef]
  180. Taha, H.B. Alzheimer’s disease and related dementias diagnosis: A biomarkers meta-analysis of general and CNS extracellular vesicles. npj Dement. 2025, 1, 3. [Google Scholar] [CrossRef]
  181. Bhatnagar, D.; Ladhe, S.; Kumar, D. Discerning the Prospects of miRNAs as a Multi-Target Therapeutic and Diagnostic for Alzheimer’s Disease. Mol. Neurobiol. 2023, 60, 5954–5974. [Google Scholar] [CrossRef] [PubMed]
  182. Da Conceicao, A.R.R.; Marinatto, J.; Pinheiro, L.S.; Rody, T.; De Felice, F.G. The Multifaceted Role of Extracellular Vesicles in Alzheimer’s Disease. J. Neurochem. 2025, 169, e70209. [Google Scholar] [CrossRef]
  183. Winston, C.N.; Goetzl, E.J.; Schwartz, J.B.; Elahi, F.M.; Rissman, R.A. Complement protein levels in plasma astrocyte-derived exosomes are abnormal in conversion from mild cognitive impairment to Alzheimer’s disease dementia. Alzheimer’s Dement. 2019, 11, 61–66. [Google Scholar] [CrossRef]
  184. TaŞDelen, E.; Kizil, E.T.Ö.; AydemİR, S.T.; Yalap, Ö.E.; BİNgÖL, A.P.; Kutlay, N. Determination of miR-373 and miR-204 levels in neuronal exosomes in Alzheimer?s disease. Turk. J. Med. Sci. 2022, 52, 1458–1467. [Google Scholar] [CrossRef]
  185. Tian, C.; Stewart, T.; Hong, Z.; Guo, Z.; Aro, P.; Soltys, D.; Pan, C.; Peskind, E.R.; Zabetian, C.P.; Shaw, L.M.; et al. Blood extracellular vesicles carrying synaptic function- and brain-related proteins as potential biomarkers for Alzheimer’s disease. Alzheimer’s Dement. 2022, 19, 909–923. [Google Scholar] [CrossRef] [PubMed]
  186. Jia, L.; Zhu, M.; Kong, C.; Pang, Y.; Zhang, H.; Qiu, Q.; Wei, C.; Tang, Y.; Wang, Q.; Li, Y.; et al. Blood neuro-exosomal synaptic proteins predict Alzheimer’s disease at the asymptomatic stage. Alzheimer’s Dement. 2021, 17, 49–60. [Google Scholar] [CrossRef]
  187. Zhao, A.; Li, Y.; Yan, Y.; Qiu, Y.; Li, B.; Xu, W.; Wang, Y.; Liu, J.; Deng, Y. Increased prediction value of biomarker combinations for the conversion of mild cognitive impairment to Alzheimer’s dementia. Transl. Neurodegener. 2020, 9, 30. [Google Scholar] [CrossRef]
  188. Kumar, A.; Su, Y.; Sharma, M.; Singh, S.; Kim, S.; Peavey, J.J.; Suerken, C.K.; Lockhart, S.N.; Whitlow, C.T.; Craft, S.; et al. MicroRNA expression in extracellular vesicles as a novel blood-based biomarker for Alzheimer’s disease. Alzheimer’s Dement. 2023, 19, 4952–4966. [Google Scholar] [CrossRef] [PubMed]
  189. Li, Y.; Xia, M.; Meng, S.; Wu, D.; Ling, S.; Chen, X.; Liu, C. MicroRNA-29c-3p in dual-labeled exosome is a potential diagnostic marker of subjective cognitive decline. Neurobiol. Dis. 2022, 171, 105800. [Google Scholar] [CrossRef] [PubMed]
  190. Durur, D.Y.; Tastan, B.; Ugur Tufekci, K.; Olcum, M.; Uzuner, H.; Karakulah, G.; Yener, G.; Genc, S. Alteration of miRNAs in Small Neuron-Derived Extracellular Vesicles of Alzheimer’s Disease Patients and the Effect of Extracellular Vesicles on Microglial Immune Responses. J. Mol. Neurosci. 2022, 72, 1182–1194. [Google Scholar] [CrossRef]
  191. Witwer, K.W.; Wolfram, J. Extracellular vesicles versus synthetic nanoparticles for drug delivery. Nat. Rev. Mater. 2021, 6, 103–106. [Google Scholar] [CrossRef]
  192. Elsharkasy, O.M.; Nordin, J.Z.; Hagey, D.W.; de Jong, O.G.; Schiffelers, R.M.; Andaloussi, S.E.; Vader, P. Extracellular vesicles as drug delivery systems: Why and how? Adv. Drug. Deliv. Rev. 2020, 159, 332–343. [Google Scholar] [CrossRef]
  193. Patil, S.M.; Sawant, S.S.; Kunda, N.K. Exosomes as drug delivery systems: A brief overview and progress update. Eur. J. Pharm. Biopharm. 2020, 154, 259–269. [Google Scholar] [CrossRef]
  194. Bruno, S.; Collino, F.; Deregibus, M.C.; Grange, C.; Tetta, C.; Camussi, G. Microvesicles derived from human bone marrow mesenchymal stem cells inhibit tumor growth. Stem Cells Dev. 2013, 22, 758–771. [Google Scholar] [CrossRef]
  195. Xia, Y.; Zhang, J.; Liu, G.; Wolfram, J. Immunogenicity of Extracellular Vesicles. Adv. Mater. 2024, 36, e2403199. [Google Scholar] [CrossRef]
  196. Han, G.; Zhang, Y.; Zhong, L.; Wang, B.; Qiu, S.; Song, J.; Lin, C.; Zou, F.; Wu, J.; Yu, H.; et al. Generalizable anchor aptamer strategy for loading nucleic acid therapeutics on exosomes. EMBO Mol. Med. 2024, 16, 1027–1045. [Google Scholar] [CrossRef]
  197. Pang, C.; Zhang, J.; Gu, Y.; Zhang, Q.; Zhao, Y. The biological roles of exosome-encapsulated traditional Chinese medicine monomers in neuronal disorders. J. Pharm. Anal. 2025, 15, 101131. [Google Scholar] [CrossRef] [PubMed]
  198. Shaffi, C.S.; Hairuddin, O.N.; Mansor, S.F.; Syafiq, T.M.F.; Yahaya, B.H. Unlocking the Potential of Extracellular Vesicles as the Next Generation Therapy: Challenges and Opportunities. Tissue Eng. Regen. Med. 2024, 21, 513–527. [Google Scholar] [CrossRef] [PubMed]
  199. Liu, X.; Lu, S.; Wan, M.; Xie, H.; Wang, Z. Peripheral extracellular vesicles in neurodegeneration: Pathogenic influencers and therapeutic vehicles. J. Nanobiotechnol. 2024, 22, 170. [Google Scholar] [CrossRef] [PubMed]
  200. Skotland, T.; Sagini, K.; Sandvig, K.; Llorente, A. An emerging focus on lipids in extracellular vesicles. Adv. Drug Deliv. Rev. 2020, 159, 308–321. [Google Scholar] [CrossRef]
  201. Williams, A.; Branscome, H.; Kashanchi, F.; Batrakova, E.V. Targeting of Extracellular Vesicle-Based Therapeutics to the Brain. Cells 2025, 14, 548. [Google Scholar] [CrossRef]
  202. Grange, C.; Tapparo, M.; Bruno, S.; Chatterjee, D.; Quesenberry, P.J.; Tetta, C.; Camussi, G. Biodistribution of mesenchymal stem cell-derived extracellular vesicles in a model of acute kidney injury monitored by optical imaging. Int. J. Mol. Med. 2014, 33, 1055–1063. [Google Scholar] [CrossRef]
  203. Qin, Q.; Shan, Z.; Xing, L.; Jiang, Y.; Li, M.; Fan, L.; Zeng, X.; Ma, X.; Zheng, D.; Wang, H.; et al. Synergistic effect of mesenchymal stem cell-derived extracellular vesicle and miR-137 alleviates autism-like behaviors by modulating the NF-kappaB pathway. J. Transl. Med. 2024, 22, 446. [Google Scholar] [CrossRef] [PubMed]
  204. Macyczko, J.R.; Wang, N.; Zhao, J.; Ren, Y.; Lu, W.; Ikezu, T.C.; Zhao, N.; Liu, C.C.; Bu, G.; Li, Y. Suppression of Wnt/beta-Catenin Signaling Is Associated with Downregulation of Wnt1, PORCN, and Rspo2 in Alzheimer’s Disease. Mol. Neurobiol. 2023, 60, 26–35. [Google Scholar] [CrossRef]
  205. Tapia-Rojas, C.; Inestrosa, N.C. Loss of canonical Wnt signaling is involved in the pathogenesis of Alzheimer’s disease. Neural Regen. Res. 2018, 13, 1705–1710. [Google Scholar]
  206. Wang, Q.; Huang, X.; Su, Y.; Yin, G.; Wang, S.; Yu, B.; Li, H.; Qi, J.; Chen, H.; Zeng, W.; et al. Activation of Wnt/beta-catenin pathway mitigates blood-brain barrier dysfunction in Alzheimer’s disease. Brain 2022, 145, 4474–4488. [Google Scholar] [CrossRef]
  207. Warrier, S.; Marimuthu, R.; Sekhar, S.; Bhuvanalakshmi, G.; Arfuso, F.; Das, A.K.; Bhonde, R.; Martins, R.; Dharmarajan, A. sFRP-mediated Wnt sequestration as a potential therapeutic target for Alzheimer’s disease. Int. J. Biochem. Cell Biol. 2016, 75, 104–111. [Google Scholar] [CrossRef]
  208. Vargas, J.Y.; Fuenzalida, M.; Inestrosa, N.C. In vivo activation of Wnt signaling pathway enhances cognitive function of adult mice and reverses cognitive deficits in an Alzheimer’s disease model. J. Neurosci. 2014, 34, 2191–2202. [Google Scholar] [CrossRef]
  209. Varshini, M.S.; Reddy, R.A.; Krishnamurthy, P.T.; Wadhwani, A. Harmony of Wnt pathway in Alzheimer’s: Navigating the multidimensional progression from preclinical to clinical stages. Neurosci. Biobehav. Rev. 2024, 165, 105863. [Google Scholar] [CrossRef] [PubMed]
  210. Li, C.; Jiao, G.; Wu, W.; Wang, H.; Ren, S.; Zhang, L.; Zhou, H.; Liu, H.; Chen, Y. Exosomes from Bone Marrow Mesenchymal Stem Cells Inhibit Neuronal Apoptosis and Promote Motor Function Recovery via the Wnt/beta-catenin Signaling Pathway. Cell Transplant. 2019, 28, 1373–1383. [Google Scholar] [CrossRef]
  211. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/beta-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  212. Zhang, H.; Wu, X.; Zhang, X.; Sun, Y.; Yan, Y.; Shi, H.; Zhu, Y.; Wu, L.; Pan, Z.; Zhu, W.; et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/beta-catenin pathway. Stem Cells Transl. Med. 2015, 4, 513–522. [Google Scholar] [CrossRef] [PubMed]
  213. Chen, Y.; Li, J.; Ma, B.; Li, N.; Wang, S.; Sun, Z.; Xue, C.; Han, H.; Wei, J.; Zhao, R.C. MSC-derived exosomes promote recovery from traumatic brain injury via microglia/macrophages in rat. Aging 2020, 12, 18274–18296. [Google Scholar] [CrossRef]
  214. Dolen, D.; Ahmadov, T.; Dolas, I.; Unal, T.C.; Aydoseli, A.; Ozturk, M.; Sabanci, P.A.; Aras, Y.; Bilgic, M.B.; Sencer, A. Analysis of the Prognosis of High-Grade gliomas in the View of New Immunohistochemistry Markers and 2016 WHO Classification. Turk. Neurosurg. 2022, 32, 500–507. [Google Scholar] [CrossRef]
  215. Zuo, R.; Liu, M.; Wang, Y.; Li, J.; Wang, W.; Wu, J.; Sun, C.; Li, B.; Wang, Z.; Lan, W.; et al. BM-MSC-derived exosomes alleviate radiation-induced bone loss by restoring the function of recipient BM-MSCs and activating Wnt/beta-catenin signaling. Stem Cell Res. Ther. 2019, 10, 30. [Google Scholar] [CrossRef]
  216. Yu, A.; Zhang, Y.; Zhong, S.; Yang, Z.; Xie, M. Human umbilical cord mesenchymal stem cell-derived exosomes enhance follicular regeneration in androgenetic alopecia via activation of Wnt/beta-catenin pathway. Stem Cell Res. Ther. 2025, 16, 418. [Google Scholar] [CrossRef]
  217. Xie, X.; Song, Q.; Dai, C.; Cui, S.; Tang, R.; Li, S.; Chang, J.; Li, P.; Wang, J.; Li, J.; et al. Clinical safety and efficacy of allogenic human adipose mesenchymal stromal cells-derived exosomes in patients with mild to moderate Alzheimer’s disease: A phase I/II clinical trial. Gen. Psychiatry 2023, 36, e101143. [Google Scholar] [CrossRef]
  218. Morita, J.; Hirano, A.; Izawa, H. Safety and clinical efficacy on intranasal administration of mesenchymal stem cell-derived secretome in patients with Alzheimer’s disease and its future prospect. Glycative Stress Res. 2024, 11, 103–110. [Google Scholar]
  219. AEGLE Therapeutics. Available online: https://aegletherapeutics.com/ev-therapy/ (accessed on 30 December 2025).
  220. Codiak BioSciences, Inc. Available online: https://www.biospace.com/codiak-presents-preclinical-data-on-exoaso-stat6-and-exoaso-c-ebp%CE%B2-programs-at-the-society-for-immunotherapy-of-cancer-sitc-2022-annual-meeting (accessed on 30 December 2025).
  221. Therapeutics, E. Available online: https://www.evoxtherapeutics.com/pipeline/ (accessed on 30 December 2025).
  222. Serrano, D.R.; Juste, F.; Anaya, B.J.; Ramirez, B.I.; Sanchez-Guirales, S.A.; Quispillo, J.M.; Hernandez, E.M.; Simon, J.A.; Trallero, J.M.; Serrano, C.; et al. Exosome-Based Drug Delivery: A Next-Generation Platform for Cancer, Infection, Neurological and Immunological Diseases, Gene Therapy and Regenerative Medicine. Pharmaceutics 2025, 17, 1336. [Google Scholar] [CrossRef] [PubMed]
  223. EXO Biologics. Available online: https://exoxpert.com/exoxpert-an-exosome-cdmo-launched-by-exo-biologics/?utm_source=chatgpt.com (accessed on 30 December 2025).
  224. Li, Y.; Chen, Y.H.; Liu, B.Y.; Nie, Q.; Li, L.J.; Duan, X.; Wu, L.Z.; Chen, G. Deciphering the Heterogeneity Landscape of Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles for Precise Selection in Translational Medicine. Adv. Healthc. Mater. 2023, 12, e2202453. [Google Scholar] [CrossRef]
  225. Ioannou, S.; Kay, A.G.; Stone, A.P.; Rand, E.; Elberfeld, S.; Bolton, W.; Larson, T.; Crossland, R.E.; Kehoe, O.; Mentlak, D.A.; et al. Extracellular vesicle bioactivity and potential for clinical development are determined by mesenchymal stromal cell clonal subtype. Stem Cell Res. Ther. 2025, 16, 571. [Google Scholar] [CrossRef]
  226. Hagey, D.W.; Ojansivu, M.; Bostancioglu, B.R.; Saher, O.; Bost, J.P.; Gustafsson, M.O.; Gramignoli, R.; Svahn, M.; Gupta, D.; Stevens, M.M.; et al. The cellular response to extracellular vesicles is dependent on their cell source and dose. Sci. Adv. 2023, 9, eadh1168. [Google Scholar] [CrossRef]
  227. Ahn, S.H.; Ryu, S.W.; Choi, H.; You, S.; Park, J.; Choi, C. Manufacturing Therapeutic Exosomes: From Bench to Industry. Mol. Cells 2022, 45, 284–290. [Google Scholar] [CrossRef] [PubMed]
  228. Konoshenko, M.Y.; Lekchnov, E.A.; Vlassov, A.V.; Laktionov, P.P. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. BioMed Res. Int. 2018, 2018, 8545347. [Google Scholar] [CrossRef] [PubMed]
  229. Andriolo, G.; Provasi, E.; Brambilla, A.; Panella, S.; Soncin, S.; Cicero, V.L.; Radrizzani, M.; Turchetto, L.; Barile, L. Methodologies for Scalable Production of High-Quality Purified Small Extracellular Vesicles from Conditioned Medium. Methods Mol. Biol. 2023, 2668, 69–98. [Google Scholar]
  230. Visan, K.S.; Lobb, R.J.; Ham, S.; Lima, L.G.; Palma, C.; Edna, C.P.Z.; Wu, L.Y.; Gowda, H.; Datta, K.K.; Hartel, G.; et al. Comparative analysis of tangential flow filtration and ultracentrifugation, both combined with subsequent size exclusion chromatography, for the isolation of small extracellular vesicles. J. Extracell. Vesicles 2022, 11, 12266. [Google Scholar] [CrossRef]
  231. Huang, J.; Chen, H.; Li, N.; Liu, P.; Yang, J.; Zhao, Y. Emerging technologies towards extracellular vesicles large-scale production. Bioact. Mater. 2025, 52, 338–365. [Google Scholar] [CrossRef]
  232. Murphy, D.E.; de Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef] [PubMed]
  233. Massa, M.; Croce, S.; Campanelli, R.; Abba, C.; Lenta, E.; Valsecchi, C.; Avanzini, M.A. Clinical Applications of Mesenchymal Stem/Stromal Cell Derived Extracellular Vesicles: Therapeutic Potential of an Acellular Product. Diagnostics 2020, 10, 999. [Google Scholar] [CrossRef]
  234. Chu, M.; Wang, H.; Bian, L.; Huang, J.; Wu, D.; Zhang, R.; Fei, F.; Chen, Y.; Xia, J. Nebulization Therapy with Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes for COVID-19 Pneumonia. Stem Cell Rev. Rep. 2022, 18, 2152–2163. [Google Scholar] [CrossRef]
  235. Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef]
  236. Fang, S.; Tian, H.; Li, X.; Jin, D.; Li, X.; Kong, J.; Yang, C.; Yang, X.; Lu, Y.; Luo, Y.; et al. Clinical application of a microfluidic chip for immunocapture and quantification of circulating exosomes to assist breast cancer diagnosis and molecular classification. PLoS ONE 2017, 12, e0175050. [Google Scholar] [CrossRef]
  237. Wang, Z.; Wu, H.J.; Fine, D.; Schmulen, J.; Hu, Y.; Godin, B.; Zhang, J.X.; Liu, X. Ciliated micropillars for the microfluidic-based isolation of nanoscale lipid vesicles. Lab Chip 2013, 13, 2879–2882. [Google Scholar] [CrossRef]
  238. He, M.; Crow, J.; Roth, M.; Zeng, Y.; Godwin, A.K. Integrated immunoisolation and protein analysis of circulating exosomes using microfluidic technology. Lab Chip 2014, 14, 3773–3780. [Google Scholar] [CrossRef] [PubMed]
  239. Zhao, Z.; Yang, Y.; Zeng, Y.; He, M. A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. Lab Chip 2016, 16, 489–496. [Google Scholar] [CrossRef]
  240. Park, E.J.; Zhang, Y.Z.; Vykhodtseva, N.; McDannold, N. Ultrasound-mediated blood-brain/blood-tumor barrier disruption improves outcomes with trastuzumab in a breast cancer brain metastasis model. J. Control Release 2012, 163, 277–284. [Google Scholar] [CrossRef] [PubMed]
  241. Lipsman, N.; Meng, Y.; Bethune, A.J.; Huang, Y.; Lam, B.; Masellis, M.; Herrmann, N.; Heyn, C.; Aubert, I.; Boutet, A.; et al. Blood-brain barrier opening in Alzheimer’s disease using MR-guided focused ultrasound. Nat. Commun. 2018, 9, 2336. [Google Scholar] [CrossRef]
  242. Wu, L.; Lin, B.; Fan, H.; Li, Y. Establishment of an ultrasound-responsive microfluidic chip BBB-glioblastoma model for studying sonodynamic therapy-enhanced nanodrug delivery. Lab Chip 2025, 25, 6673–6687. [Google Scholar] [CrossRef] [PubMed]
  243. Leinenga, G.; Götz, J. Scanning ultrasound removes amyloid-b and restores memory in an Alzheimer’s disease mouse model. Sci. Transl. Med. 2015, 7, 278ra33. [Google Scholar] [CrossRef]
  244. Shen, Y.; Hua, L.; Yeh, C.K.; Shen, L.; Ying, M.; Zhang, Z.; Liu, G.; Li, S.; Chen, S.; Chen, X.; et al. Ultrasound with microbubbles improves memory, ameliorates pathology and modulates hippocampal proteomic changes in a triple transgenic mouse model of Alzheimer’s disease. Theranostics 2020, 10, 11794–11819. [Google Scholar] [CrossRef]
  245. Rezai, A.R.; D’Haese, P.F.; Finomore, V.; Carpenter, J.; Ranjan, M.; Wilhelmsen, K.; Mehta, R.I.; Wang, P.; Najib, U.; Vieira Ligo Teixeira, C.; et al. Ultrasound Blood-Brain Barrier Opening and Aducanumab in Alzheimer’s Disease. N. Engl. J. Med. 2024, 390, 55–62. [Google Scholar] [CrossRef]
  246. Jordao, J.F.; Thevenot, E.; Markham-Coultes, K.; Scarcelli, T.; Weng, Y.Q.; Xhima, K.; O’Reilly, M.; Huang, Y.; McLaurin, J.; Hynynen, K.; et al. Amyloid-beta plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound. Exp. Neurol. 2013, 248, 16–29. [Google Scholar] [CrossRef]
  247. Deng, Z.; Wang, J.; Xiao, Y.; Li, F.; Niu, L.; Liu, X.; Meng, L.; Zheng, H. Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-beta-induced neurotoxicity. Theranostics 2021, 11, 4351–4362. [Google Scholar] [CrossRef] [PubMed]
  248. Ye, D.; Chen, S.; Liu, Y.; Weixel, C.; Hu, Z.; Yuan, J.; Chen, H. Mechanically manipulating glymphatic transport by ultrasound combined with microbubbles. Proc. Natl. Acad. Sci. USA 2023, 120, e2212933120. [Google Scholar] [CrossRef]
Figure 1. The dual role of EVs in AD pathogenesis. The schematic illustrates the opposing functions of EVs in AD, contrasting their pathological role in propagating the disease (top, “Dark side”) with their endogenous protective and therapeutic functions (bottom, “Bright side”). On the dark side, EVs derived from neurons and glial cells act as mediators of AD progression by facilitating the intercellular propagation of Aβ and tau pathologies, exacerbating neuroinflammation through the releasing of pro-inflammatory factors, and disseminating oxidative stress. Conversely, on the bright side, EVs-from endogenous neurons, glial cells and mesenchymal stem cells (MSC-EVs)-orchestrate a multifaceted defense by promoting the clearance of pathogenic Aβ and tau, delivering neuroprotective and immunomodulatory cargo to suppress inflammation, preserving synaptic integrity, alleviating oxidative stress and modulating brain metabolism.
Figure 1. The dual role of EVs in AD pathogenesis. The schematic illustrates the opposing functions of EVs in AD, contrasting their pathological role in propagating the disease (top, “Dark side”) with their endogenous protective and therapeutic functions (bottom, “Bright side”). On the dark side, EVs derived from neurons and glial cells act as mediators of AD progression by facilitating the intercellular propagation of Aβ and tau pathologies, exacerbating neuroinflammation through the releasing of pro-inflammatory factors, and disseminating oxidative stress. Conversely, on the bright side, EVs-from endogenous neurons, glial cells and mesenchymal stem cells (MSC-EVs)-orchestrate a multifaceted defense by promoting the clearance of pathogenic Aβ and tau, delivering neuroprotective and immunomodulatory cargo to suppress inflammation, preserving synaptic integrity, alleviating oxidative stress and modulating brain metabolism.
Pharmaceutics 18 00070 g001
Figure 2. Future perspectives: Integrated EV-based platforms for AD diagnosis and therapy. Schematic overview of two promising technologies for the diagnosis and treatment of AD. Top panel: Microfluidic chip for blood-based EVs diagnostics. This platform outlines a “liquid biopsy” workflow for AD. Plasma is introduced into a microfluidic chip where NDEVs are selectively immunocaptured using antibody against neuronal surface markers (e.g., L1CAM). The captured exosomes are subsequently lysed, and their cargo (e.g., Aβ, p-tau, synaptic proteins) is quantitatively analyzed via on-chip immunoassays, enabling minimally invasive diagnosis and disease monitoring. Bottom panel: Focused Ultrasound to enhance EV therapy. A non-invasive FUS device transiently opens the BBB, intrinsically facilitates Aβ clearance, and restore cognitive function. This physical disruption enhanced the delivery of therapeutic EVs—including MSC-EVs, engineered EVs (e.g., loaded with CRISPR/Cas9 systems or anti-inflammatory miRNAs) and plant-derived EVs (e.g., broccoli-derived exosomes)—to the brain parenchyma, amplifying their therapeutic effect on core AD pathologies.
Figure 2. Future perspectives: Integrated EV-based platforms for AD diagnosis and therapy. Schematic overview of two promising technologies for the diagnosis and treatment of AD. Top panel: Microfluidic chip for blood-based EVs diagnostics. This platform outlines a “liquid biopsy” workflow for AD. Plasma is introduced into a microfluidic chip where NDEVs are selectively immunocaptured using antibody against neuronal surface markers (e.g., L1CAM). The captured exosomes are subsequently lysed, and their cargo (e.g., Aβ, p-tau, synaptic proteins) is quantitatively analyzed via on-chip immunoassays, enabling minimally invasive diagnosis and disease monitoring. Bottom panel: Focused Ultrasound to enhance EV therapy. A non-invasive FUS device transiently opens the BBB, intrinsically facilitates Aβ clearance, and restore cognitive function. This physical disruption enhanced the delivery of therapeutic EVs—including MSC-EVs, engineered EVs (e.g., loaded with CRISPR/Cas9 systems or anti-inflammatory miRNAs) and plant-derived EVs (e.g., broccoli-derived exosomes)—to the brain parenchyma, amplifying their therapeutic effect on core AD pathologies.
Pharmaceutics 18 00070 g002
Table 2. Blood-based EVs functioning as diagnostic biomarkers for AD.
Table 2. Blood-based EVs functioning as diagnostic biomarkers for AD.
EV SubtypeSourceChanges in the Components of EVsDiseaseConclusionReferences
NDEVsNeuron42, T-tau, and p-tau T181 ↑AD
aMCI
Blood exosomal Aβ42, T-tau, and p-tau T181 show strong concordance with CSF and offer comparable diagnostic power for AD/aMCI.[174]
42AD
aMCI
NDEVs levels of Aβ42 increase progressively across the AD continuum—from cognitively normal amyloid-negative individuals to amyloid-positive individuals, to those with MCI.[175]
42, T-tau, p-tau T181, p-tau S396 ↑AD
FID
NDEVs levels of p-tau T181, p-tau S396, and Aβ1–42 achieves 96.4% accuracy in classifying AD patients and predict disease onset 10 years prior to clinical diagnosis.[176]
p-tau T181,p-tau S396 ↑
NRGN, EST ↓
AD
MCI
The combined profile of p-tau, Aβ1–42, NRGN and REST in plasma NDEVs serves as an accurate predictor for the conversion from MCI to AD dementia.[38]
1–42
NRGN, synaptophysin ↓
synaptotagmin, synaptopodin ↓
MCI1–42, NRGN, synaptophysin, synapsin, and synaptopodin in NDEVs accurately differentiate patients MCI from CNC.[177]
p-tau T181, Aβ42, NRGN ↑
ProBDNF, GluR2, PSD95 ↓
GAP43, Syntaxin-1 ↓
ADA model incorporating GluR2, proBDNF, NRGN, and GAP43 achieved an 81.3% accuracy for AD classification. Their levels correlated with cognitive scores (MMSE and COR-SOB), supporting utility for both diagnosis and progression monitoring.[178]
miR-373, miR-204 ↓ADThe significant reduction in miR-204 and miR-373 in NDEVs positions them as potential biomarkers for AD.[184]
NMDAR2AADAnalysis of synaptic protein profiles in CNS-derived plasma EVs provides a robust, liquid biopsy biomarker for synaptic dysfunction on AD[185]
GAP43, neurogranin, SNAP25, synaptotagmin1 ↓ADNDEV proteins levels (GAP43) correlate with CSF, serving as effective biomarkers and enabling the prediction of AD onset 5 to 7 years prior to the emergence of cognitive impairment.[186]
1–42AD
MCI
The combination of Aβ1–42 levels in NDEVs accurately predicts conversion from MCI to AD dementia.[187]
miR-9-5p, miR-106b-5p, miR-125b-5p ↑AD
MCI
MCI-AD
MiR-106b-5p is significantly overexpressed in the AD group and shows perfect accuracy in distinguish AD from cognitive normal (CN) group.[188]
Aβ42/40, Tau, P-tau-T181, Aβ42, miR-29c-3p ↑SCD
aMCI
AD
VaD
The level of miR-29c-3p in NCAM/amphiphysin 1 dual-labeled exosomes (NDEVs) of patients with subjective cognitive decline (SCD) is higher than that in healthy controls and the vascular dementia (VaD) group, and it demonstrates the best performance in diagnosing SCD, holding potential advantages for early diagnosis. The biomarkers such as Aβ42 in single-labeled exosomes have diagnostic value for aMCI and AD, but limited diagnostic value for SCD.[189]
let-7e-5p, miR-96-5p, miR-484 ↑
miR-99b-5p, miR-100-5p, miR-30e-5p ↓ miR-378i, miR-145-5p, miR-378c, miR-451a ↓
ADCompared with the HC, plasma levels of sNDEV let-7e-5p, miR-96-5p, and miR-484 were significantly increased in AD group, while levels of miR-99b-5p, miR-100-5p, miR-30e-5p et al. were significantly decreased. Let-7e expressed in NDEVs could serve as a potential biomarker for AD diagnosis (AUC value of 0.9214).[190]
ADEVsAstrocyteBACE-1, g-secretase, soluble Aβ42, sAPPβ↓ sAPPa, GDNF, P-tau T181, p-tau S396 ↑AD
FID
The levels of BACE-1 and aAPPβ in ADEVs can effectively distinguish AD patients from healthy controls, with AUC values of 0.78 and 0.83, respectively.[65]
C1q, C4b, factor D, fragment ↑
Bb, C5b, C3b, C5b-C9 ↑
CD46, CD59, type 1 complement receptor ↓
MCIThe changes in complement protein levels in ADEVs not only distinguish AD patients from the control group but also demonstrate high diagnostic sensitivity in identifying MCI patients at high risk of converting to AD.[183]
miR-29a-5p, miR-132-5p, miR-107 ↑AD
MCI
MCI-AD
The expression of miR-107 shows an increasing trend among the three patient groups and can perfectly predict the incidence of AD dementia (AUC = 1.000).[188]
MDEVsMicrogliamiR-132-5p, miR-106b-5p ↑
miR-29a-5p, miR-125b-5p ↓
AD
MCI
MCI-AD
The levels of miR-29a-5p and miR-106-5p is significantly reduced across all impairment groups (AUC = 0.925). MiR-132-5p and miR-125b-5p together can perfectly predict AD (AUC = 1.000).[188]
ODEVsOligodendrocytesmiR-29a-5p, miR-107, miR-135b-5p ↑AD
MCI
MCI-AD
miR-29a-5p, miR-107, and miR-135b-5p were significantly overexpressed in the AD group. Mir-29a-5p showed AUC = 1.000 in predicting AD incidence.[188]
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

Li, F.; Wu, L.; Feng, X.; Li, Y.; Fan, H. Extracellular Vesicles in Alzheimer’s Disease: Dual Roles in Pathogenesis, Promising Avenues for Diagnosis and Therapy. Pharmaceutics 2026, 18, 70. https://doi.org/10.3390/pharmaceutics18010070

AMA Style

Li F, Wu L, Feng X, Li Y, Fan H. Extracellular Vesicles in Alzheimer’s Disease: Dual Roles in Pathogenesis, Promising Avenues for Diagnosis and Therapy. Pharmaceutics. 2026; 18(1):70. https://doi.org/10.3390/pharmaceutics18010070

Chicago/Turabian Style

Li, Feng, Liyang Wu, Xin Feng, Yihong Li, and Huadong Fan. 2026. "Extracellular Vesicles in Alzheimer’s Disease: Dual Roles in Pathogenesis, Promising Avenues for Diagnosis and Therapy" Pharmaceutics 18, no. 1: 70. https://doi.org/10.3390/pharmaceutics18010070

APA Style

Li, F., Wu, L., Feng, X., Li, Y., & Fan, H. (2026). Extracellular Vesicles in Alzheimer’s Disease: Dual Roles in Pathogenesis, Promising Avenues for Diagnosis and Therapy. Pharmaceutics, 18(1), 70. https://doi.org/10.3390/pharmaceutics18010070

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