Skip to main content

Peripheral extracellular vesicles in neurodegeneration: pathogenic influencers and therapeutic vehicles

Abstract

Neurodegenerative diseases (NDDs) such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis epitomize a class of insidious and relentless neurological conditions that are difficult to cure. Conventional therapeutic regimens often fail due to the late onset of symptoms, which occurs well after irreversible neurodegeneration has begun. The integrity of the blood-brain barrier (BBB) further impedes efficacious drug delivery to the central nervous system, presenting a formidable challenge in the pharmacological treatment of NDDs. Recent scientific inquiries have shifted focus toward the peripheral biological systems, investigating their influence on central neuropathology through the lens of extracellular vesicles (EVs). These vesicles, distinguished by their ability to breach the BBB, are emerging as dual operatives in the context of NDDs, both as conveyors of pathogenic entities and as prospective vectors for therapeutic agents. This review critically summarizes the burgeoning evidence on the role of extracerebral EVs, particularly those originating from bone, adipose tissue, and gut microbiota, in modulating brain pathophysiology. It underscores the duplicity potential of peripheral EVs as modulators of disease progression and suggests their potential as novel vehicles for targeted therapeutic delivery, positing a transformative impact on the future landscape of NDD treatment strategies.

Search strategy A comprehensive literature search was conducted using PubMed, Web of Science, and Scopus from January 2000 to December 2023. The search combined the following terms using Boolean operators: “neurodegenerative disease” OR “Alzheimer’s disease” OR “Parkinson’s disease” OR “Amyotrophic lateral sclerosis” AND “extracellular vesicles” OR “exosomes” OR “outer membrane vesicles” AND “drug delivery systems” AND “blood-brain barrier”. MeSH terms were employed when searching PubMed to refine the results. Studies were included if they were published in English, involved human subjects, and focused on the peripheral origins of EVs, specifically from bone, adipose tissue, and gut microbiota, and their association with related diseases such as osteoporosis, metabolic syndrome, and gut dysbiosis. Articles were excluded if they did not address the role of EVs in the context of NDDs or did not discuss therapeutic applications. The titles and abstracts of retrieved articles were screened using a dual-review process to ensure relevance and accuracy. The reference lists of selected articles were also examined to identify additional relevant studies.

Introduction

Neurodegenerative diseases (NDDs) epitomize a group of maladies that insidiously dismantle the central nervous system (CNS), heralding significant biomedical challenges in an aging population. These disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), not only precipitate profound neurological impairment but also impart considerable socioeconomic strain as life expectancies elongate globally [1]. NDDs are typified by the inexorable loss of neuronal integrity and function, culminating in a spectrum of symptoms from cognitive decline to motor and muscular debilitation. Traditional paradigms attribute these phenotypes to an array of cellular malfunctions, including compromised mitochondrial efficacy, oxidative stress, and defective autophagic processes [2,3,4,5,6]. Yet, these canonical pathways do not wholly delineate the complexity of NDD pathologies, nor do they provide the panacea for their cure, suggesting that additional, intricate mechanisms are at play.

At the vanguard of emerging mechanisms are extracellular vesicles (EVs), nano- to micron-sized carriers, which have garnered attention for their role in mediating cellular communication beyond the confines of the CNS [7]. These vesicles, encompassing both exosomes (30–100 nm) derived from endosomal compartments and the more heterogeneous ectosomes (50–1000 nm), encapsulate a microcosm of the cellular interior, carrying nucleic acids, proteins, lipids, and metabolites across biological frontiers [8,9,10]. Their repertoire extends beyond mere waste disposal to orchestrating physiological and pathological processes, facilitated by their capacity to breach the blood-brain barrier (BBB) and to mediate crosstalk between the CNS and peripheral tissues [7, 8, 11,12,13].

This review aims to elucidate the emerging narrative of extracerebral-derived EVs as dual agents: modulators of NDD progression and potential vectors for therapeutic intervention. By analyzing the influence of peripheral sources such as bone, adipose tissue, and gut microbiota, we aim to explain how EVs reflect and potentially initiate the molecular disruption that is characteristic of NDDs (Fig. 1). This review will traverse the landscape of recent discoveries that implicate EV-mediated peripheral-brain axis perturbations in the pathogenesis of AD, PD, and ALS. Furthermore, we will explore the broader implications of systemic illnesses that may potentiate neurodegeneration. The review will culminate in a discourse on the prospective utility of EVs in modulating brain pathology and their potential as novel therapeutic options for NDDs. In synthesizing these domains, our objective is to provide a comprehensive prospect that integrates the peripheral environmental factors with central pathologies, offering a novel lens through which to view and combat NDDs. By shedding light on the peripheral origins of EVs and their potential to traverse the BBB, we spotlight a frontier in NDD research for exploration and therapeutic innovation.

Fig. 1
figure 1

The integrative role of extracerebral-derived EVs in neurodegenerative disease pathogenesis and therapy. Extracerebral-derived EVs, originating from diverse peripheral sources such as bone, adipose tissue, and gut microbiota, are depicted as pivotal entities in the intercommunication between the periphery and the CNS. These vesicles possess the remarkable ability to traverse the systemic circulation, penetrate the BBB, and deliver molecular cargo that can influence NDD mechanisms. Additionally, the inherent biological characteristics of EVs present them as promising vectors for targeted therapeutic delivery to the CNS, offering a novel approach to treat NDDs. Engineered EVs, optimized for CNS targeting, could represent a paradigm shift in the modulation of pathogenic processes underlying NDDs and the administration of therapeutic agents

Evidence of peripheral contributions to NDD Pathogenesis

The pathogenesis of NDDs has been predominantly attributed to CNS intrinsic factors, with hallmark neuropathological features such as amyloid-β (Aβ) plaques, hyperphosphorylated tau, and misfolded alpha-synuclein (α-syn) driving the research narrative [3]. This CNS-centric paradigm has overshadowed the potential contributory role of peripheral processes in the pathophysiology of these disorders. However, recent evidence challenges this brain-autonomous perspective, underscoring the influence of peripheral systems in both the production and aggregation of neurotoxic proteins within the CNS environment.

The interplay between the peripheral environment and the brain is emerging as a significant factor in the etiopathogenesis of NDDs. Peripheral cells have been shown to secrete exogenous toxic protein species that can spread to the CNS, potentially instigating or exacerbating neurodegenerative cascades [14]. Moreover, systemic inflammation and peripheral metabolic dysfunctions are increasingly recognized as key modulators of CNS pathology, suggesting that peripheral homeostatic alterations might be upstream events in NDD development [15]. The hypothesis that peripheral-derived factors contribute to NDDs extends beyond the simple translocation of pathogenic proteins. It encompasses a more complex scenario where peripheral dysregulation, mediated through EVs and other molecular conduits, orchestrates a detrimental cross-talk that may precipitate or perpetuate neurodegeneration [16]. These insights into the peripheral origins of brain pathology represent a paradigm shift, implicating a bi-directional axis of communication between the CNS and peripheral tissues that could underlie the progression of NDDs.

Consequently, the implication of the periphery in NDDs is reshaping our understanding of these diseases. This realization opens novel avenues for the identification of peripheral biomarkers and presents a frontier for therapeutic interventions that extend beyond the confines of the CNS. The investigation into peripheral contributions thus holds the potential to redefine therapeutic strategies, emphasizing the importance of a systemic approach in combatting neurodegeneration.

Alzheimer’s disease and systemic biological interactions

AD, characterized by a profound decline in cognitive faculties and the most prevalent form of dementia, presents a formidable challenge to the medical community [17]. The histopathological signatures of AD include the extracellular deposition of Aβ peptides and the intracellular aggregation of hyperphosphorylated tau protein [18]. Despite extensive research, therapeutic strategies targeting these neuropathological features, specifically aimed at the clearance of Aβ or tau, have not yet resulted in significant clinical benefit [19]. This therapeutic impasse has prompted a reassessment of the etiopathogenic mechanisms underlying AD.

A seminal advance in this context is the growing appreciation of the systemic nature of AD, recognizing that the disease may not solely originate within the brain’s precincts. It has been posited that peripheral cells are capable of producing Aβ, which could instigate or exacerbate cerebral amyloidosis [20, 21]. This was compellingly demonstrated in a murine model where the transplantation of bone marrow cells from transgenic mice harboring AD-associated mutations into wild type (WT) counterparts resulted in the manifestation of AD-like neuropathology and cognitive deficits [22].

Moreover, the biological interplay between the periphery and the CNS is increasingly implicated in the progression of AD pathology, particularly through the roles of EVs. EVs, as carriers of intercellular communication, have been identified in the plasma of AD patients at elevated levels. They transport not only the secretases involved in APP processing but also an array of inflammatory mediators [23, 24]. This compelling evidence suggests a mechanism whereby systemic EVs may actively contribute to the neuropathological cascade of AD. Furthermore, peripheral organs exposed to aging, metal ion dysregulation, or infectious stimuli may launch an inflammatory response and release protease-rich EVs into the circulatory system. These EVs may traverse the BBB via the choroid plexus, becoming inadvertent couriers of pathogenicity to the hippocampal neurons, the epicenter of memory formation in the brain. Additionally, it has also been observed that circulating EVs in AD patients can disrupt the integrity of the BBB by diminishing the expression of vascular endothelial-cadherin, a key adherens junction protein [25]. This perturbation of the BBB may potentiate the influx of pathogenic molecules into the brain parenchyma, further fueling the neurodegenerative process.

In light of these findings, the relationship between systemic biology and AD emerges as a complex web of interactions, with implications that transcend the brain’s boundaries. By recognizing the peripheral contributions to AD, a new perspective is fostered, one that views AD not merely as a disorder of the brain but as a reflection of systemic imbalance. This systemic perspective could herald novel diagnostic markers and therapeutic targets, ultimately reshaping the strategies for combating this pervasive disease.

Parkinson’s disease: beyond the central Dogma

PD, the senescence-associated scourge of the dopaminergic system, ranks as the second most prevalent neurodegenerative disorder among the elderly [26]. The pathognomonic feature of PD is the presence of Lewy bodies, and neuronal cytoplasmic inclusions predominantly composed of misfolded α-synuclein (α-syn), which mark the neuronal landscape of the disease [27]. The classical view of PD as a disease confined to the CNS has been challenged by revelations of widespread α-syn deposition, not just in the brain but also in peripheral tissues including the intestine, pancreas, heart, salivary glands, and skin [28,29,30].

The transmissibility of α-syn pathology represents a paradigm shift in understanding the pathogenesis of PD, where EVs emerge as potential vectors for the dissemination of pathological proteins. Recent studies have implicated EVs as a mechanism for the propagation of α-syn, suggesting a prion-like model of spread within and possibly beyond the confines of the CNS [31, 32]. The role of inflammation, an ever-present specter in neurodegenerative conditions, is now being reinterpreted in the context of PD. There is accumulating evidence that neuroinflammation, arising peripherally, can potentiate central inflammatory responses, leading to the activation of microglia and atrophy of astrocytes. This cascade not only accentuates the neurodegenerative process but also magnifies the deleterious impact of α-syn oligomers, further compromising neuronal integrity and amplifying motor and non-motor deficits in PD models [33].

The involvement of peripheral EVs as conduits for inflammatory signals provides a mechanistic link between systemic inflammation and neurodegeneration. Experiments demonstrating that blood-transformed EVs from aged mice can provoke glial activation in the brains of younger recipients underscore the profound influence of systemic factors on central pathologies [34]. Moreover, EVs derived from inflamed macrophages (Mϕ) have been shown to contribute to the progression of PD pathology, intensifying neuroinflammation, and exacerbating disease pathology [35]. This body of evidence propels the notion that PD is not an isolated central phenomenon but a systemic ailment with significant peripheral contributions.

Amyotrophic lateral sclerosis and peripheral system cross-talk

ALS, an inexorably progressive neurodegenerative disorder, induces motor neuron demise, leading to pervasive muscle weakness and ultimately, respiratory failure [36]. Central neuroinflammation is recognized as a cardinal pathological feature, contributing to the relentless neuronal attrition observed in ALS [37]. Notably, the neuroinflammatory cascade in ALS is not isolated to the CNS; it involves a dynamic interplay with systemic immune components. Proinflammatory cells such as monocytes and T lymphocytes, along with their secretory immunoreactive molecules, have been implicated in disease exacerbation, indicating a substantial peripheral contribution to disease dynamics [38,39,40].

The peripheral immune signature of ALS patients has been found to be distinctly aberrant, with an altered monocyte distribution that not only reflects systemic immune dysregulation but also correlates with their capacity to breach the CNS barriers and propagate neuroinflammation [38]. This is further compounded by documented perturbations in the integrity of the CNS barriers in both ALS rodent models and patients, which potentiate the ingress of noxious peripheral factors into the neural environment [41].

EVs are emerging as pivotal entities in the molecular crosstalk between peripheral systems and the CNS in ALS. They are suspected couriers of pathogenic cargo, with a putative role in the dissemination of neurotoxic elements toward motor neurons (MNs) [40, 42]. A recent finding accentuates the toxic impact of skeletal muscle-derived EVs from sporadic ALS patients on MNs, underscoring the direct pathogenic conduits between affected muscles and neuronal cells [43]. Additionally, the therapeutic potential of EVs has been glimpsed in studies where EVs from peripheral mesenchymal stromal cells exert immunomodulatory effects, possibly mediated by microRNAs targeting microglial cells, thus offering a beacon of hope in the modulation of neuroinflammation in ALS [44].

In summary, the evidence delineating the role of peripheral factors in the pathogenesis of NDDs is compelling, warranting a re-evaluation of traditional CNS-centric models. By incorporating a holistic view that integrates systemic physiology with neuropathology, we stand on the cusp of unveiling the multifactorial nature of these complex disorders, potentially heralding a new era in their management.

Deciphering the influence of bone-derived EVs on neurodegeneration

The skeletal system, an often-overlooked contributor to neurodegenerative pathology, dispatches EVs into the peripheral circulation that bears the capacity to influence brain health [45]. Bone-derived EVs, encompassing those from bone marrow stem cells, immune cells, and osteocytes, are increasingly recognized for their role in modulating neural environments, with potential implications in the pathogenesis of NDDs. In the nexus between bone health and neurodegeneration, current research delineates a neuroprotective axis steered by the release of EVs from healthy bone marrow stem cells. These EVs are armed with regenerative cargo, growth factors, anti-inflammatory cytokines, and neurotrophic molecules, that endorse neuronal survival and plasticity [46, 47]. The dichotomy in the role of bone marrow immune cell-derived EVs is stark; while some subsets foster neuroprotection and repair, others may convey pro-inflammatory signals, exacerbating neurodegenerative processes [48, 49].

Recent epidemiological threads weave a compelling narrative; diseases deteriorating bone integrity, such as osteoporosis (OP) and osteoarthritis (OA), are correlated with an augmented risk of AD and PD [47, 50,51,52,53,54]. This interrelationship hints at a scenario where diseased bone could pivot from a sanctuary to a source of pathological EVs. Dysfunctional osteocyte-derived EVs, in the environment of diseases such as OP, may lose their neuroprotective attributes or, worse, transport pathological molecules that could precipitate or aggravate NDDs [47].

Osteo-neural crosstalk: EVs from bone marrow stem cells

The intercommunication between the bone marrow niche and the CNS is a complex and bidirectional dialogue, in which bone marrow-derived stem cells (BMSCs) including hematopoietic stem cells (HSCs), mesenchymal stem cells (BM-MSCs), and endothelial progenitor cells (EPCs), play pivotal roles. These cells are endowed with the remarkable plasticity to self-renew and differentiate into various cell types, thus standing at the forefront of regenerative medicine and neuroprotection [55, 56]. HSCs are instrumental in CNS repair mechanisms, primarily through trans-differentiation processes. Long-standing evidence intimates a compromised regenerative capacity in the context of NDDs; specifically, an attenuated hematopoietic supplement is observed in patients with AD and PD [57, 58]. This insufficiency predicates a deleterious impact on the intrinsic repair capabilities of CNS. BM-MSCs have garnered substantial attention due to their therapeutic potential in neurodegenerative disorders. Notably, intravenous administration of BM-MSCs has been shown to mitigate pathological alterations and foster neurogenesis within murine models of AD and PD, evincing their regenerative abilities [55, 59, 60]. Furthermore, the intrathecal introduction of these cells in a rodent model of ALS has led to significant disease modification, enhancing motor function and prolonging lifespan [61].

The therapeutic effects of BMSCs may be attributed, at least in part, to the EVs they secrete. These EVs possess the functional components derived from their progenitor cells, carrying inherent targeting properties and the capacity to transverse biological barriers [62, 63]. In AD murine models, BM-MSC-derived EVs, upon intracerebral administration, have exhibited efficacy in reducing Aβ plaque burdens and neuronal atrophy, notably within the cerebral cortex and hippocampus [64]. These vesicles carry the upregulation of brain-derived neurotrophic factor (BDNF) and neprilysin (NEP), a critical Aβ-degrading enzyme, simultaneously attenuating neuroinflammatory processes [62, 65]. Human BM-MSC-derived EVs have further demonstrated their capacity to induce differentiation in neural progenitor cells and have yielded beneficial outcomes in PD animal models, enhancing both motor function and histological integrity [66]. Additionally, EPC-derived EVs have been documented to interface with brain endothelial cells, promoting the restoration of the compromised blood-CNS barrier in ALS models [67, 68].

Immuno-regulatory role of bone marrow immune cells-derived EVs

The bone marrow serves as a reservoir for the immune cells, producing a diverse array of components integral to the function of immune system [69]. Among these, EVs derived from bone marrow immune cells have emerged as crucial mediators of intercellular communication, influencing both physiological and pathological processes, including neurodegeneration. Bone marrow immune cells, encompassing a broad spectrum from myeloid progenitors to mature monocytes and lymphocytes, actively secrete EVs that participate in the regulation of immune responses [49]. These vesicles carry a payload of bioactive molecules, including cytokines, growth factors, and nucleic acids, that can modulate immune activity within the CNS [70]. The perturbation of EV signaling may contribute to the pathogenic processes underlying NDDs, suggesting a potential therapeutic target or biomarker for disease progression.

The crosstalk between the peripheral immune system and the CNS is a critical component of neuroinflammatory responses in neurodegeneration [71, 72]. EVs from bone marrow immune cells can cross the BBB, influencing the CNS environment. They are implicated in various mechanisms, such as modulating the activation state of microglia, the resident immune cells of the brain, which have been associated with the pathogenesis of diseases like multiple sclerosis (MS) and AD [73, 74]. The pathophysiological relevance of myeloid-derived EVs in NDDs is underscored by their capability to influence neuroinflammation. In models of AD, for instance, myeloid-derived EVs have been shown to enhance the phagocytic activity of microglia towards Aβ aggregates, indicating a potential for therapeutic modulation of innate immune responses [75].

As biomarkers, bone marrow-derived EVs hold promise for the early diagnosis and monitoring of NDDs. Their presence and alterations in their molecular cargo in the peripheral blood may reflect CNS pathology, allowing for less invasive assessments of disease state and progression [70]. Future studies must aim to elucidate the precise molecular mechanisms governing EV-mediated interactions between the bone marrow and CNS to harness their full therapeutic and diagnostic potential.

Osteocytes as remote regulators of CNS health

Osteocytes, the chief architects of the bone microenvironment, emerge as unconcerned conductors in the maintenance of CNS health, underscoring a novel paradigm in neurobiology. These cells, constituting up to 95% of the bone matrix populace, boast remarkable longevity, with their lifespan extending over two decades, echoing the protracted durability of neurons. Osteocytes extend an intricate network of dendritic processes (on average 50), similar to neuronal dendrites, facilitating intercellular communication through gap junctions and canalicular conduits within the mineralized bone matrix [76]. Historically relegated to the role of mechanical placeholders within bone, osteocytes are now recognized as dynamic endocrine players, orchestrating bone remodeling and mineral metabolism [77]. Beyond their traditional confines, osteocytes reach into systemic physiology through their secretome, influencing distal organ functions [76, 78, 79]. Their ability to dispatch EVs containing bioactive molecules underscores a remote regulatory capacity that extends to the CNS.

Emergent evidence delineates the role of osteocyte-derived EVs as conveyors of neuroprotection. Our investigations unveiled that osteocytes harvested from young mice secrete EVs that potentiate the mitigation of cognitive decline and interfere with the pathological cascade of AD [47]. These findings pivotally suggest that osteocytes, through their secreted EVs, may exert a salutary influence on the CNS, providing a potential therapeutic option. The age-related decline in the regenerative capacity of osteocyte-derived EVs reflects a parallel with neurodegenerative progression, suggesting a senescence-associated diminishment in their neuroprotective efficacy. The characterization of osteocyte-derived EVs from young versus aged sources reveals differential molecular signatures, indicating a potential shift from neurotrophic to neuroinflammatory profiles with aging. This posits osteocyte-derived EVs as both biomarkers and modifiable entities in the context of neurodegeneration. Investigations into the mechanistic pathways engaged by osteocyte-derived EVs highlight their role in modulating neuroinflammatory responses, amyloidogenic pathways, and synaptic plasticity. Through the delivery of specific proteins and signaling molecules, these vesicles may attenuate neuroinflammatory cascades and modulate Aβ metabolism, offering insights into the crosstalk between skeletal health and cognitive function.

Recognition of osteocytes as peripheral regulators of CNS health via EV-mediated signaling has revealed a previously underappreciated dimension of the bone-brain axis. The prospect of leveraging the neuroprotective potential of osteocyte-derived EVs opens new avenues for therapeutic intervention in NDDs, potentially altering the trajectory of conditions such as AD. As the research landscape evolves, the imperative to decode the molecular language of osteocyte-derived EVs will be paramount in translating these findings into clinical reality.

In summary, emerging evidence suggest that bone derived EVs as important messengers within the bone-brain axis, integral to the maintenance of cerebral integrity (Fig. 2). These vesicular entities, dispatched by various cellular constituents of the bone, including marrow stem cells, immune cells, and osteocytes, harbor the capacity to transduce signals conducive to neuroprotection and cognitive function preservation. However, it is observed that the neuroregulatory potential of bone-derived EVs declines in the face of osteocellular senescence or pathological states such as OP, aligning with an increased vulnerability to NDDs. The attrition of osteocellular vitality, whether through apoptotic demise or dormancy, appears to coincide with a decline in the supportive secretome, implicating bone health as a critical factor in the etiopathogenesis of NDDs [80,81,82]. This inverse correlation between the robustness of bone-derived EVs and the incidence of disorders such as AD and PD illuminates the nuanced interplay between systemic skeletal conditions and neural degeneration. As such, osteopathy may not merely coexist with neurodegeneration but may actively precipitate its onset and progression, underscoring a need to re-evaluate therapeutic targets within this axis.

Fig. 2
figure 2

Deciphering the influence of bone-derived EVs on neurodegeneration. Bone derived EVs serve as important messengers in the bone-brain axis. EVs derived from bone marrow stem cells, immune cells, and osteocytes carry an array of functional molecules, including various proteins and miRNAs, which are implicated in neuroprotective mechanisms. These benefits are reduced when cells in the bones die or become inactive due to aging or disease states

Adipose tissue-derived EVs: messengers of metabolic and neurologic health

The interplay between adipose tissue and the CNS extends far beyond mere energy storage, engaging in a complex dialogue critical for maintaining metabolic and neurologic homeostasis [83]. As the primary energy reservoir and an endocrine powerhouse, adipose tissue orchestrates a symphony of hormones and bioactive molecules, profoundly influencing peripheral and central processes [84]. This section elucidates the dualistic nature of adipose tissue-derived EVs as conveyors of both metabolic information and neuropathological consequences.

Dysfunction in adipose tissue, marked by altered secretion of adipokines such as leptin and adiponectin, emerges as a pivotal factor in NDD etiology [85,86,87,88]. However, despite long-term intake of hypercaloric nutrition, the energy status of brain is lowered in obese patients [89]. The paradox of decreased cerebral energy availability in the face of hypercaloric intake, as observed in obesity, underscores a metabolic disconnect that extends to compromised neurotrophic support. Notably, diets rich in fats and sugars precipitate declines in brain-derived neurotrophic factor (BDNF) expression [90, 91], which is essential for neuronal survival and synaptic integrity. Proinflammatory adipokines further exacerbate neuroinflammation, disrupting CNS homeostasis [88, 92]. In this inflammatory environment, adipose tissue-derived EVs serve as both carriers of crisis and harbingers of healing. These vesicles, harnessing the intrinsic communicative properties of adipocytes, disseminate signals that can be pathogenic or protective, depending on their molecular cargoes and the metabolic context from which they originate [93].

Among the reparative pathways activated by adipose-derived signals, MSCs within adipose depots have garnered attention. Adipose tissue-derived MSCs (AD-MSCs) and their EVs exhibit pronounced neuroprotective effects [94,95,96,97,98]. In vitro, EVs from human AD-MSCs (hAD-MSCs) mitigate microglial activation and inflammatory cytokine release, simultaneously enhancing anti-inflammatory mediator expression [96]. Moreover, in animal models of ALS, AD-MSC-derived EVs home to affected neural regions, attenuating motor neuron degeneration and glial activation [97].

However, the pathogenic potential of adipocyte-derived EVs surfaces in the context of obesity and diabetes. EVs from such compromised states can induce cognitive decline and synaptic deficits in otherwise healthy recipients, implicating miRNAs like miR-9-3p in the dysregulation of neuronal insulin signaling [99]. Furthermore, adipocyte EVs isolated from AD patients carry miRNAs (for example, miR-6760-3p and miR-6798-3p) that may perturb the cyclic AMP response element binding protein (CREB) signaling pathway in neurons, pivotal for memory and synaptic plasticity [100].

In summary, adipose tissue-derived EVs constitute a crucial nexus between metabolic and neurologic health. In a state of physiological equilibrium, they transport molecules that uphold cognitive function and neural resilience. Conversely, metabolic dysregulation yields EVs laden with deleterious factors that propagate neuroinflammatory and degenerative pathways. Figure 3 delineates this duality, emphasizing the consequential role of adipose-derived signals in the integrity of the CNS.

Fig. 3
figure 3

Adipose tissue-derived EVs: messengers of metabolic and neurologic health. EVs deriving from adipose tissue present the potential to mediate both neuroprotective and neuropathogenic interactions. In the upper panel, EVs released from healthy adipose tissue are rich in beneficial adipokines, anti-inflammatory cytokines, and growth factors that collectively support neurogenesis, synaptic plasticity, and cognitive function. In the lower panel, in the metabolic disorders pathological state, adipose tissue becomes a source of EVs laden with proinflammatory adipokines, dysregulated miRNAs, and other metabolic by-products that can impair cognitive function

.

Gut-brain axis: microbiota-derived EVs as modulators of neural integrity

The intricate ecosystem of the human gut microbiota is a pivotal contributor to homeostasis, extending its influence beyond the gastrointestinal tract to modulate immune, neurological, and metabolic functions [101]. The gut-brain axis (GBA) epitomizes this bidirectional communication, with the gut microbiota acting as a key mediator [102, 103]. Disturbances in the microbiota, or dysbiosis, triggered by factors like antibiotics, stress, or infection, disrupt this communication and may lead to a permeabilized gut barrier, allowing for the systemic dissemination of pro-inflammatory agents [104].

Emerging evidence underscores the relationship between microbial dysbiosis and NDD pathogenesis, linking microbial imbalance to CNS anomalies, such as heightened BBB permeability, neuroinflammation, and accumulation of neurotoxic proteins [102, 105]. This imbalance in homeostasis can lead to infection by opportunistic pathogenic bacteria and increased BBB permeability, thus promoting neuroinflammation and the accumulation of neurotoxic misfolded proteins in the CNS [106,107,108,109]. Conversely, probiotic treatments have shown promising results. Strains of Lactobacillus and Bifidobacterium, for instance, have been noted to ameliorate cognitive deficits in AD models [110, 111], and Akkermansia muciniphila has demonstrated potential in alleviating symptoms in neurodegenerative mouse models [112, 113]. Probiotics can also help patients with PD to result in beneficial impacts on movement disorders and constipation [114].

Of particular interest are EVs secreted by gut bacteria. These EVs are similar to yet distinct from, mammalian exosomes and ectosomes, as they form through the budding of the bacterial outer membrane [115, 116]. These bacterial EVs have emerged as novel communicators within the GBA, traversing to the host brain via circulatory or neural pathways [117, 118]. For instance, Helicobacter pylori (HP) EVs can breach the BBB and provoke neuroinflammation through mechanisms involving glial cell activation and complement pathway signaling, potentially exacerbating amyloidogenic pathology [119, 120]. Moreover, neurotoxic EVs from pathogens, like Pseudomonas aeruginosa, Paenalcaligenes hominis, and Aggregatibacter actinomycetemcomitans, contain components such as lipopolysaccharides (LPS) that can invoke a robust immune response in the CNS [115, 121, 122]. In stark contrast, EVs from commensal bacteria with probiotic characteristics offer therapeutic promise. EVs from Lactobacillus plantarum, for example, have elicited antidepressant effects and counteracted amyloid-beta induced neuropathology in experimental models [123, 124].

Thus, the modulation of the intestinal microbiota, favoring beneficial over pathogenic bacteria through dietary or pharmacological interventions, may be a viable strategy for preserving neural integrity. Probiotic-derived EVs, in particular, present an innovative avenue for therapeutic exploration, potentially offering a means to restore or maintain neurological health through the harmonization of the GBA. The clinical translation of these findings could revolutionize treatment paradigms for neurodegenerative disorders, providing a novel class of biologics derived from the gut microbiota itself (Fig. 4).

Fig. 4
figure 4

Gut-brain axis: microbiota-derived EVs as modulators of neural integrity. This schematic illustrates the dichotomous influences of EVs derived from gut microbiota on neural health. Probiotic EVs, originating from beneficial commensals, confer neuroprotective effects, which may ameliorate NDD pathology. Conversely, disruption of gut homeostasis allows pathogenic bacteria to release EVs that can translocate across the intestinal and the BBB. These vesicles transport inflammatory components, such as LPS, peptidoglycans, and bacterial toxins, into the CNS, potentially inducing immune responses that precipitate neuroinflammation and the aggregation of neurotoxic proteins

Overall, the effects of extracerebral-derived EVs on NDDs have a dual nature and can be modulated through various mechanisms. Table 1 provides a summary of the roles of different sources of extracerebral-derived EVs in NDDs, emphasizing their diverse functions and impacts on the pathogenesis of these disorders.

Table 1 Essential functions of EVs and their impact on NDDs

Therapeutic horizons: harnessing peripheral EVs for NDD intervention

In the vanguard of regenerative medicine and the therapeutic landscape for NDDs, EVs stand out as a promising cell-free modality, circumventing the complexities and ethical quandaries often associated with stem cell transplantation and other cell-based interventions [126]. These naturally occurring nanocarriers harbor the intrinsic ability to encapsulate and convey bioactive molecules—proteins, lipids, and nucleic acids—reflective of their cells of origin, thus preserving the potential for therapeutic modulation [127]. Their structural bilipid membrane composition not only ensures the integrity of their molecular cargo but also facilitates their navigation across the biological moat that is the BBB, a feature that is indispensable for the targeted delivery of therapeutics to the CNS [128].

The distinctive characteristic of EVs to serve as vectors for intercellular communication positions them as uniquely suited for the conveyance of reparative signals to neural tissues beleaguered by degenerative processes. In this paradigm, the horizon of NDD intervention is broadening, with EVs emerging as not only vehicles for direct therapeutic action but also as agents that can modulate the neuroinflammatory environment that is a hallmark of neurodegeneration. Moreover, the capacity of EVs to be engineered for enhanced targeting provides a platform for precision medicine, wherein modifications to EV surface proteins or contents can be tailored to address specific pathophysiological conditions within the CNS.

This section delineates the therapeutic potential of peripheral EVs, exploring the frontiers of their application in NDDs and discussing the mechanistic underpinnings that position these entities as a novel class of therapeutic agents. We will scrutinize the emerging evidence for their capability to modulate disease pathways, deliver pharmacological agents across the BBB, and initiate reparative processes within the diseased CNS. By harnessing the innate properties of EVs, and through innovative bioengineering strategies, there is a tantalizing prospect for the development of novel therapeutics that can navigate the challenges presented by the complexity of the human brain and the diseases that afflict it.

Navigating the BBB: the trojan horse strategy of EVs

The BBB represents a formidable selective interface that preserves the neural environment from systemic influences. Constituted by brain microvascular endothelial cells (BMECs), this dynamic barrier, in concert with pericytes and astrocytes, composes the neurovascular unit (NVU), a complex sentinel that governs the cerebral ingress and egress of molecular entities. The discerning permeability of BBB is pivotal for neural homeostasis but simultaneously poses significant impediments to the pharmacotherapy of neurodegenerative disorders, thwarting the passage of most therapeutic agents [129, 130].

However, EVs with their innate ability to traverse biological membranes emerge as a ‘Trojan Horse’, deftly navigating the BBB. Recent empirical evidence underscores the heterogeneous penetrative capacities of EVs across this barrier. A recent investigative delineated the trans-BBB trafficking of various EV populations, discerning a spectrum of permeability across mouse, human, neoplastic, and non-neoplastic cellular lineages, thereby illuminating the differential crossing efficacies that could be harnessed therapeutically [131].

The precise modalities by which EVs infiltrate the NVU remain an area of intense research. It is posited that transcellular transport of EVs ensues via endocytic uptake by BMECs, an event capable of instigating phenotypic alterations within these cells and modulating the selective permeability of BBB [25, 132, 133]. This is exemplified by studies demonstrating that inducible pluripotent stem cell-derived small EVs (iPSC-sEVs) appear to counteract senescence in the BBB, offering cytoprotection in ischemic cerebrovascular insults [134]. Further elucidating the potential of EVs, it has been shown that MSC-derived EVs, as well as those from brain endothelial origins, can fortify the integrity of the BBB by modulating the Caveolin-1-dependent trafficking of tight junction proteins, such as ZO-1 and Claudin-5, thus preserving barrier function during acute ischemic events [135].

In addition to transcellular routes, EVs may exploit paracellular pathways, transiting via the inter-endothelial junctional complexes under certain physiological conditions [128]. Conversely, pathological derangements in the BBB, such as those encountered in NDD states, may precipitate a heightened permeability that permits the ingress of neurotoxic EVs, exacerbating neuronal injury [129]. This dualistic impact of EVs on the BBB, either as vehicles of repair or as conveyors of damage, heralds a nuanced understanding of their role in CNS pathobiology.

EVs represent a versatile arsenal in the quest to overcome the pharmacological sanctuary imposed by the BBB. The ability of EVs to interface with the NVU and modulate its properties not only underscores their potential as delivery vectors for therapeutic agents but also as modifiers of barrier physiology with implications for disease modulation. Further elucidation of the mechanisms guiding EV translocation and the subsequent effects on the NVU will enhance our capacity to exploit these nanovesicles for therapeutic gain.

Biogenesis of therapeutically competent EVs: potential cellular factories

The therapeutic deployment of EVs in NDD management necessitates an intricate understanding of their biogenesis and the cellular provenance that confers them with a neuroprotective and regenerative prowess. MSCs, by their inherent propensity to secrete EVs laden with reparative molecules, stand at the forefront of potential cellular factories. Their autologous use in clinical settings harnesses this natural reservoir of therapeutic agents, offering a promising avenue for the amelioration of NDDs [63, 136].

The landscape of potential cellular sources for therapeutic EVs extends beyond MSCs. Dendritic cells (DCs), Mϕ, and neutrophils, including neutrophil-like promyelocytic cells, are increasingly recognized for their contributory roles in this context. DC-derived EVs, pivotal in the orchestration of immune responses, also partake in antigen presentation, a function that might be exploitable for disease-modifying strategies in NDDs [137,138,139]. Of particular interest are EVs from immature DCs, which are proficient in the release of EVs devoid of T-cell co-stimulatory molecules, such as major histocompatibility complex class II (MHC-II), thereby proffering an immunologically quiescent profile for therapeutic purposes [140].

Monocytes and Mϕ represent another cadre of cellular factories, capable of generating EVs that can elude the reticuloendothelial system, thereby circumventing one of the major barriers to effective drug delivery [138]. Such an attribute not only enhances the direct transport of therapeutic cargoes to targeted cells but also augments the therapeutic index of encapsulated drugs [139].

Moreover, neutrophils and their progenitors, the promyelocytes, are emerging as prolific producers of EVs. Their ability to rapidly secrete large quantities of EVs, coupled with their innate biological functions, positions them as a valuable source for the generation of EVs tailored for drug delivery. The drug-delivery systems derived from such EVs could potentially leverage the natural navigation systems of neutrophils, which instinctively migrate towards inflammation sites, a common feature of NDD pathogenesis.

Collectively, these cellular sources offer a palette of biogenetic origins for the derivation of therapeutically competent EVs. The selection of specific cellular factories for EV production must consider not only the innate characteristics of the EVs they produce but also the pathophysiological context of the NDD in question. Understanding the nuances of EV biogenesis from these diverse cellular origins will advance our capacity to tailor EV-based therapies to the complex demands of NDD intervention. Such advancements will pave the way for more precise and effective treatments, harnessing the full potential of EVs as carriers of regenerative medicine.

Bioengineering EVs for precision targeting of CNS pathology

The precise targeting of CNS pathologies using EVs represents a paradigm shift in the therapeutic approach to NDDs. However, the inherent limitations of natural EVs, namely their suboptimal targeting efficiency, limited circulation half-life, and variable therapeutic payload delivery, necessitate the application of advanced bioengineering techniques to enhance their functionality and therapeutic precision.

The modification of EV surface ligands represents a critical frontier in this effort [141]. Utilizing innovative bioengineering techniques, researchers have sought to augment the innate homing capabilities of EVs. A pioneering approach by Alvarez-Erviti et al. demonstrated the potential of such modifications, where DCs were engineered to express a chimeric protein consisting of the lysosome-associated membrane protein 2b (Lamp2b) fused with a neuron-specific rabies viral glycoprotein (RVG) peptide. The EVs resulting from this molecular alteration have a heightened affinity for neuronal tissues, subsequently increasing their accumulation in regions pivotal for neurodegenerative processes, such as the cerebral cortex and hippocampus [137]. Further refinements in this domain have been illustrated by Cui et al., who showed that MSC-derived EVs (MSC-EVs) modified with the RVG peptide not only preferentially homed to neural tissue but also conferred notable anti-inflammatory effects in an AD mouse model. These bioengineered MSC-RVG-EVs downregulated pro-inflammatory cytokines, upregulated anti-inflammatory IL-10, and importantly, improved cognitive function, which is a significant therapeutic outcome [142].

Beyond surface engineering, conditioning of parental cells under specific culture conditions has emerged as a potent strategy to influence the biological activity of secreted EVs. For instance, AD-MSCs cultured in neurogenesis-promoting media have been shown to yield EVs with enhanced neural differentiation potential, indicating that the microenvironmental conditioning of progenitor cells can significantly impact the therapeutic profile of EVs [125]. Moreover, the microenvironmental oxygen tension has been implicated in the modulation of EV function. Studies by Huang et al. revealed that AD-MSCs subjected to hypoxic conditions not only exhibited increased EV secretion but also an enriched expression of specific microRNAs like miR-511-3p, known for their anti-inflammatory properties [143]. Additionally, there has been growing interest in utilizing advanced bioreactor technologies to scale up EV production. Jeske et al. reported that culturing human MSCs (hMSCs) in a 3D dynamic bioreactor significantly boosted the yield of EVs and enhanced the profile of their therapeutic cargo, suggesting that bioprocessing strategies can be pivotal in optimizing EV-based therapies for clinical applications [144].

The cumulative advancements in the bioengineering of EVs hold the promise of creating precision-targeted therapeutic agents capable of navigating the complex biological environment of the CNS. By leveraging these sophisticated methodologies, there is a realizable prospect of developing a new class of EV-based interventions that can address the intricate challenges posed by neurodegenerative pathologies with unprecedented specificity and efficacy.

The Future of drug delivery: EVs in the clinical setting

The landscape of therapeutic strategies for NDDs is on the cusp of a significant paradigm shift with the advent of EVs as drug delivery systems (DDS) [145]. Nanotechnology has paved the way for innovative approaches aimed at traversing the complex environment of the CNS. However, the functional attributes of any DDS, including biodistribution and drug release profiles, are inherently dependent upon the physicochemical properties of the carrier system [128].

Traditional DDS such as polymeric nanoparticles and liposomes have been explored extensively, with both modalities revealing unique advantages and inherent limitations. Polymers like polycaprolactone (PCL), polylactic acid (PLA), and poly(lactic-co-glycolic acid) (PLGA), despite their versatility, are often challenged by suboptimal drug loading efficiencies and potential localized immune responses [129, 146]. Liposomes, with their biocompatible phospholipid constructs, offer advantages in drug encapsulation but are not exempt from issues like off-target effects and rapid clearance from circulation [129].

EVs, by contrast, emerge as endogenously derived vectors that are inherently biocompatible, low in immunogenicity, and proficient at crossing the BBB. This facilitates a targeted delivery mechanism that is both specific to the neural tissue and favorable in terms of reduced systemic toxicity [128, 147, 148]. The global research community is actively unraveling the potential of EVs in this domain.

The structural integrity of EVs allows for prolonged storage at cryogenic temperatures with minimal degradation, ensuring the stability of encapsulated therapeutic agents ranging from enzymes to nucleic acids [148, 149]. This has led to successful encapsulation and delivery of therapeutically active molecules like the CB2 receptor agonist AM1241, which exhibits enhanced brain bioavailability and therapeutic efficacy when delivered via MSC-EVs [150]. Similarly, bioactive compounds such as quercetin and curcumin, as well as protein modulators like tyrosine phosphatase-2 (SHP2), have been efficiently packaged within EVs, providing a therapeutic thrust against hyperphosphorylated tau and mitochondrial dysfunction associated with AD [151,152,153]. For PD, catalase and dopamine have been loaded in EVs to attenuate neuroinflammation and replenish neurotransmitter levels respectively [138, 154].

RNA-based therapeutics have garnered interest for their capacity to modify disease progression. Engineered EVs carrying BACE1-targeted siRNA have demonstrated a potent capacity to mitigate Aβ synthesis by significantly suppressing BACE1 expression in vivo [137, 155]. Further, miRNA-enriched EVs have been shown to exert neuroprotective effects, with miR-29 and miR-22 variants downregulating pathological Aβ production and inflammatory cascades in AD models [156, 157]. Additionally, m6A modifications in RNA processes, implicated in PD, have been targeted using MSC-EVs to deliver demethylase FTO-targeted siRNAs, demonstrating neuroprotective outcomes [158].

We have encapsulated this burgeoning field in Table 2, summarizing the prospective therapeutic EVs, their cellular origins, and their bioactive constituents. Mirroring the swift advancements in mRNA-based therapeutics for infectious diseases and oncology, the prospective integration of mRNA drugs into EV platforms for NDD treatment holds immense promise [159]. This burgeoning frontier of EV-based mRNA delivery could potentially redefine the therapeutic approach to NDDs, offering a vehicle of unprecedented specificity and reduced immunogenicity in the clinical armamentarium against these intractable conditions.

Table 2 Summary of EVs administration for NDDs

Conclusion and future prospects

In conclusion, this review provides a comprehensive overview of the dual role of extracerebral-derived EVs in the CNS and NDDs. We present new insights that, on the one hand, extracerebral-derived EVs have a protective effect on the brain in healthy states. On the other hand, with the occurrence of aging or disease, EVs derived from the periphery carry deleterious molecules that mediate disruption of the periphery-brain axis, which leads to the development of brain disorders such as NDDs (Fig. 5). This review has endeavored to distill the dichotomous nature of extracerebral-derived EVs within the neural landscape, elucidating their paradoxical roles as both custodians of cerebral integrity and potential harbingers of neurodegeneration. We have detailed the emerging paradigm where, in a state of homeostasis, extracerebral EVs contribute to the maintenance of brain health. In contrast, the advent of aging or pathologic states precipitates a shift, wherein these same vesicles become conveyors of detrimental cargo, effectuating dysregulation of the periphery-brain axis and potentially inciting the onset of NDDs (Fig. 5). Simultaneously, this narrative has spotlighted peripheral cell-secreted EVs as a novel and promising therapeutic modality for NDDs. On the one hand, the generation or release of harmful extracerebral EVs can be effectively suppressed through the use of specific protein inhibitors. One such inhibitor is the hyaluronic acid synthetase inhibitor, which has been shown to block the uploading of these detrimental EVs [23]. By treating the underlying peripheral diseases that drive the secretion of these harmful EVs, their secretion can also be significantly reduced. On the other hand, the secretion of beneficial EVs can be specifically stimulated or augmented. For instance, mechanical loading has been shown to revitalize osteocytes and enhance the secretion of EVs derived from them [164, 165]. This suggests that beneficial EVs may have the potential to be further optimized and their therapeutic efficacy enhanced through advanced engineering techniques. By harnessing these strategies simultaneously, it may be possible to achieve more significant therapeutic outcomes

Fig. 5
figure 5

Role of extracerebral-derived EVs in pathogenesis of NDDs. This schematic delineates the dualistic influence of extracerebral-derived EVs on the progression of NDDs. In physiologically normal conditions, EVs originating from peripheral tissues such as bone, adipose, and the gut microbiome are instrumental in safeguarding cerebral function. Conversely, during pathological states like OP, metabolic syndrome, and gut dysbiosis, these extracerebral EVs facilitate a perturbation of the peripheral-brain axis. Such disturbances are implicated in neuronal attrition, degenerative changes, accumulation of neurotoxic species, and heightened neuroinflammatory responses, cumulatively exacerbating the pathogenesis of NDDs

Despite their potential, the trajectory toward the clinical application of EVs is fraught with complexities. The current landscape is limited by the absence of uniform protocols for EV isolation and a comprehensive understanding of their biogenetic and secretory machinery. To advance from bench to bedside, a rigorous framework for the optimization of EV-producing cell types is necessitated. This involves refining methodologies for cultivation, purification, and precise characterization of EVs. Moreover, it is imperative to undertake extensive safety evaluations, specifically in the context of long-term implications. The path to therapeutic implementation is one of incremental advancements. With each step, we gain deeper insights into the molecular mechanism of EV function and their interaction with neurobiological systems. In the future, it is envisioned that the integration of nanotechnology, bioengineering, and molecular biology will yield innovative platforms for the targeted delivery of EV-based therapies. In parallel, advances in omics technologies and systems biology promise to unravel the molecular signatures of EVs, fostering the development of bespoke therapeutic strategies

The prospective utility of EVs as vectors for precision medicine in NDDs holds immense promise. It beckons a new era of intervention strategies that leverage the innate biological synergy between EVs and the CNS. To actualize this potential, interdisciplinary collaboration and sustained investment in research are essential. The evolution of regulatory frameworks to accommodate these novel entities will be equally critical. As we chart this unexplored territory, the future beckons with the promise of harnessing the full therapeutic arsenal of EVs to confront the insidious challenge of NDDs.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019;15:565–81.

    Article  PubMed  Google Scholar 

  2. Wang Y, Xu E, Musich PR, Lin F. Mitochondrial dysfunction in neurodegenerative diseases and the potential countermeasure. CNS Neurosci Ther. 2019;25:816–24.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Vaquer-Alicea J, Diamond MI. Propagation of protein aggregation in neurodegenerative diseases. Annu Rev Biochem. 2019;88:785–810.

    Article  CAS  PubMed  Google Scholar 

  4. Teleanu DM, Niculescu AG, Lungu II, Radu CI, Vladacenco O, Roza E, et al. An overview of oxidative stress, Neuroinflammation, and neurodegenerative diseases. Int J Mol Sci. 2022;23:5938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chen ML, Hong CG, Yue T, Li HM, Duan R, Hu WB, et al. Inhibition of mir-331-3p and mir-9-5p ameliorates Alzheimer’s disease by enhancing autophagy. Theranostics. 2021;11:2395–409.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Liu Z, Li H, Hong C, Chen M, Yue T, Chen C, et al. ALS-Associated E478G mutation in human OPTN (Optineurin) promotes inflammation and induces neuronal cell death. Front Immunol. 2018;9:2647.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Xiao Y, Wang SK, Zhang Y, Rostami A, Kenkare A, Casella G, et al. Role of extracellular vesicles in neurodegenerative diseases. Prog Neurobiol. 2021;201:102022.

    Article  PubMed  Google Scholar 

  8. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015;16:24–43.

    Article  CAS  PubMed  Google Scholar 

  10. Mathieu M, Martin-Jaular L, Lavieu G, Thery C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21:9–17.

    Article  CAS  PubMed  Google Scholar 

  11. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.

    Article  PubMed  Google Scholar 

  12. Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, Pereira de Almeida L. Extracellular vesicles: novel promising delivery systems for therapy of brain diseases. J Control Release. 2017;262:247–58.

    Article  CAS  PubMed  Google Scholar 

  13. Liu X, Chen C, Jiang Y, Wan M, Jiao B, Liao X, et al. Brain-derived extracellular vesicles promote bone-fat imbalance in Alzheimer’s disease. Int J Biol Sci. 2023;19:2409–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Castillo X, Castro-Obregon S, Gutierrez-Becker B, Gutierrez-Ospina G, Karalis N, Khalil AA, et al. Re-thinking the Etiological Framework of Neurodegeneration. Front Neurosci. 2019;13:728.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Jorfi M, Maaser-Hecker A, Tanzi RE. The neuroimmune axis of Alzheimer’s disease. Genome Med. 2023;15:6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gulen MF, Samson N, Keller A, Schwabenland M, Liu C, Gluck S, et al. cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature. 2023;620:374–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kozlov S, Afonin A, Evsyukov I, Bondarenko A. Alzheimer’s disease: as it was in the beginning. Rev Neurosci. 2017;28:825–43.

    Article  CAS  PubMed  Google Scholar 

  18. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chetelat G, Teunissen CE, et al. Alzheimer’s disease. Lancet. 2021;397:1577–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Livingston G, Sommerlad A, Orgeta V, Costafreda SG, Huntley J, Ames D, et al. Dementia prevention, intervention, and care. Lancet. 2017;390:2673–734.

    Article  PubMed  Google Scholar 

  20. Bu XL, Xiang Y, Jin WS, Wang J, Shen LL, Huang ZL, et al. Blood-derived amyloid-beta protein induces Alzheimer’s disease pathologies. Mol Psychiatry. 2018;23:1948–56.

    Article  CAS  PubMed  Google Scholar 

  21. Kuo YM, Kokjohn TA, Watson MD, Woods AS, Cotter RJ, Sue LI, et al. Elevated abeta42 in skeletal muscle of Alzheimer disease patients suggests peripheral alterations of AbetaPP metabolism. Am J Pathol. 2000;156:797–805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sun HL, Chen SH, Yu ZY, Cheng Y, Tian DY, Fan DY, et al. Blood cell-produced amyloid-beta induces cerebral Alzheimer-type pathologies and behavioral deficits. Mol Psychiatry. 2021;26:5568–77.

    Article  CAS  PubMed  Google Scholar 

  23. Lee JH, Ostalecki C, Oberstein T, Schierer S, Zinser E, Eberhardt M, et al. Alzheimer’s disease protease-containing plasma extracellular vesicles transfer to the hippocampus via the choroid plexus. EBioMedicine. 2022;77:103903.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Ben Khedher MR, Haddad M, Fulop T, Laurin D, Ramassamy C. Implication of circulating extracellular vesicles-bound amyloid-beta42 oligomers in the progression of Alzheimer’s disease. J Alzheimers Dis. 2023;96:813–825.

  25. Bei J, Miranda-Morales EG, Gan Q, Qiu Y, Husseinzadeh S, Liew JY, et al. Circulating exosomes from Alzheimer’s Disease suppress vascular endothelial-cadherin expression and induce barrier dysfunction in recipient brain microvascular endothelial cell. J Alzheimers Dis. 2023;95:869–85.

    Article  CAS  PubMed  Google Scholar 

  26. Bloem BR, Okun MS, Klein C. Parkinson’s disease. Lancet. 2021;397:2284–303.

    Article  CAS  PubMed  Google Scholar 

  27. Jankovic J, Tan EK. Parkinson’s disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020;91:795–808.

    Article  PubMed  Google Scholar 

  28. Adler CH, Dugger BN, Hentz JG, Hinni ML, Lott DG, Driver-Dunckley E, et al. Peripheral synucleinopathy in early Parkinson’s Disease: Submandibular Gland Needle Biopsy findings. Mov Disord. 2016;31:250–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gibbons CH, Garcia J, Wang N, Shih LC, Freeman R. The diagnostic discrimination of cutaneous alpha-synuclein deposition in Parkinson disease. Neurology. 2016;87:505–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Titova N, Padmakumar C, Lewis SJG, Chaudhuri KR. Parkinson’s: a syndrome rather than a disease? J Neural Transm (Vienna). 2017;124:907–14.

    Article  CAS  PubMed  Google Scholar 

  31. Yang Y, Stewart T, Zhang C, Wang P, Xu Z, Jin J et al. Erythrocytic alpha-synuclein and the gut microbiome: kindling of the gut-brain axis in Parkinson’s disease. Mov Disord. 2024;39:40–52.

  32. Stuendl A, Kunadt M, Kruse N, Bartels C, Moebius W, Danzer KM, et al. Induction of alpha-synuclein aggregate formation by CSF exosomes from patients with Parkinson’s disease and dementia with Lewy bodies. Brain. 2016;139:481–94.

    Article  PubMed  Google Scholar 

  33. La Vitola P, Balducci C, Baroni M, Artioli L, Santamaria G, Castiglioni M, et al. Peripheral inflammation exacerbates alpha-synuclein toxicity and neuropathology in Parkinson’s models. Neuropathol Appl Neurobiol. 2021;47:43–60.

    Article  PubMed  Google Scholar 

  34. Morales-Prieto DM, Murrieta-Coxca JM, Stojiljkovic M, Diezel C, Streicher PE, Henao-Restrepo JA, et al. Small extracellular vesicles from Peripheral blood of aged mice pass the blood-brain barrier and induce glial cell activation. Cells. 2022;11:625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Jin Y, Wu R, Li L, Shen L, Gu Y, Sun C. Exosomes from Inflamed macrophages promote the progression of Parkinson’s disease by inducing Neuroinflammation. Mol Neurobiol. 2023;60:1914–28.

    Article  CAS  PubMed  Google Scholar 

  36. Feldman EL, Goutman SA, Petri S, Mazzini L, Savelieff MG, Shaw PJ, et al. Amyotrophic lateral sclerosis. Lancet. 2022;400:1363–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Beers DR, Appel SH. Immune dysregulation in amyotrophic lateral sclerosis: mechanisms and emerging therapies. Lancet Neurol. 2019;18:211–20.

    Article  CAS  PubMed  Google Scholar 

  38. Zondler L, Muller K, Khalaji S, Bliederhauser C, Ruf WP, Grozdanov V, et al. Peripheral monocytes are functionally altered and invade the CNS in ALS patients. Acta Neuropathol. 2016;132:391–411.

    Article  CAS  PubMed  Google Scholar 

  39. Chiot A, Zaidi S, Iltis C, Ribon M, Berriat F, Schiaffino L, et al. Modifying macrophages at the periphery has the capacity to change microglial reactivity and to extend ALS survival. Nat Neurosci. 2020;23:1339–51.

    Article  CAS  PubMed  Google Scholar 

  40. Carata E, Muci M, Di Giulio S, Mariano S, Panzarini E. Looking to the future of the role of macrophages and extracellular vesicles in neuroinflammation in ALS. Int J Mol Sci. 2023;24:11251.

  41. Liu Z, Cheng X, Zhong S, Zhang X, Liu C, Liu F, et al. Peripheral and central nervous system Immune Response Crosstalk in Amyotrophic lateral sclerosis. Front Neurosci. 2020;14:575.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Upadhya R, Zingg W, Shetty S, Shetty AK. Astrocyte-derived extracellular vesicles: neuroreparative properties and role in the pathogenesis of neurodegenerative disorders. J Control Release. 2020;323:225–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Le Gall L, Duddy WJ, Martinat C, Mariot V, Connolly O, Milla V, et al. Muscle cells of sporadic amyotrophic lateral sclerosis patients secrete neurotoxic vesicles. J Cachexia Sarcopenia Muscle. 2022;13:1385–402.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Giunti D, Marini C, Parodi B, Usai C, Milanese M, Bonanno G, et al. Role of miRNAs shuttled by mesenchymal stem cell-derived small extracellular vesicles in modulating neuroinflammation. Sci Rep. 2021;11:1740.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Frame G, Bretland KA, Dengler-Crish CM. Mechanistic complexities of bone loss in Alzheimer’s disease: a review. Connect Tissue Res. 2020;61:4–18.

    Article  PubMed  Google Scholar 

  46. Zhang J, Buller BA, Zhang ZG, Zhang Y, Lu M, Rosene DL, et al. Exosomes derived from bone marrow mesenchymal stromal cells promote remyelination and reduce neuroinflammation in the demyelinating central nervous system. Exp Neurol. 2022;347:113895.

    Article  CAS  PubMed  Google Scholar 

  47. Jiang YL, Wang ZX, Liu XX, Wan MD, Liu YW, Jiao B et al. The Protective Effects of Osteocyte-Derived Extracellular Vesicles Against Alzheimer’s Disease Diminished with Aging. Adv Sci (Weinh). 2022:e2105316.

  48. Zhou R, Wang L, Chen L, Feng X, Zhou R, Xiang P et al. Bone Marrow-Derived GCA(+) Immune Cells Drive Alzheimer’s Disease Progression. Adv Sci (Weinh). 2023:e2303402.

  49. Makhijani P, McGaha TL. Myeloid responses to Extracellular vesicles in Health and Disease. Front Immunol. 2022;13:818538.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Sheng Z, Xu K, Ou Y, Dai R, Luo X, Liu S, et al. Relationship of body composition with prevalence of osteoporosis in central south Chinese postmenopausal women. Clin Endocrinol (Oxf). 2011;74:319–24.

    Article  PubMed  Google Scholar 

  51. Pi YZ, Wu XP, Liu SP, Luo XH, Cao XZ, Xie H, et al. Age-related changes in bone biochemical markers and their relationship with bone mineral density in normal Chinese women. J Bone Min Metab. 2006;24:380–5.

    Article  CAS  Google Scholar 

  52. Zhou R, Zhou H, Rui L, Xu J. Bone loss and osteoporosis are associated with conversion from mild cognitive impairment to Alzheimer’s disease. Curr Alzheimer Res. 2014;11:706–13.

    Article  CAS  PubMed  Google Scholar 

  53. Kwon MJ, Kim JH, Kim JH, Cho SJ, Nam ES, Choi HG. The occurrence of Alzheimer’s Disease and Parkinson’s disease in individuals with osteoporosis: a Longitudinal Follow-Up study using a National Health Screening database in Korea. Front Aging Neurosci. 2021;13:786337.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Innes KE, Sambamoorthi U. The Association of Osteoarthritis and Related Pain Burden to Incident Alzheimer’s Disease and related dementias: a retrospective cohort study of U.S. Medicare Beneficiaries. J Alzheimers Dis. 2020;75:789–805.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Wu CC, Wang IF, Chiang PM, Wang LC, Shen CJ, Tsai KJ. G-CSF-mobilized bone marrow mesenchymal stem cells replenish neural lineages in Alzheimer’s Disease mice via CXCR4/SDF-1 Chemotaxis. Mol Neurobiol. 2017;54:6198–212.

    Article  CAS  PubMed  Google Scholar 

  56. Cao J, Jin L, Yan ZQ, Wang XK, Li YY, Wang Z, et al. Reassessing endothelial-to-mesenchymal transition in mouse bone marrow: insights from lineage tracing models. Nat Commun. 2023;14:8461.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Maler JM, Spitzer P, Lewczuk P, Kornhuber J, Herrmann M, Wiltfang J. Decreased circulating CD34 + stem cells in early Alzheimer’s disease: evidence for a deficient hematopoietic brain support? Mol Psychiatry. 2006;11:1113–5.

    Article  CAS  PubMed  Google Scholar 

  58. Funk N, Wieghofer P, Grimm S, Schaefer R, Buhring HJ, Gasser T, et al. Characterization of peripheral hematopoietic stem cells and monocytes in Parkinson’s disease. Mov Disord. 2013;28:392–5.

    Article  PubMed  Google Scholar 

  59. Ahmed HH, Salem AM, Atta HM, Eskandar EF, Farrag AR, Ghazy MA, et al. Updates in the pathophysiological mechanisms of Parkinson’s disease: emerging role of bone marrow mesenchymal stem cells. World J Stem Cells. 2016;8:106–17.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Kaniowska D, Wenk K, Rademacher P, Weiss R, Fabian C, Schulz I, et al. Extracellular vesicles of mesenchymal stromal cells can be taken up by Microglial cells and partially prevent the Stimulation Induced by beta-amyloid. Stem Cell Rev Rep. 2022;18:1113–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Forostyak S, Homola A, Turnovcova K, Svitil P, Jendelova P, Sykova E. Intrathecal delivery of mesenchymal stromal cells protects the structure of altered perineuronal nets in SOD1 rats and amends the course of ALS. Stem Cells. 2014;32:3163–72.

    Article  CAS  PubMed  Google Scholar 

  62. Liu S, Fan M, Xu JX, Yang LJ, Qi CC, Xia QR, et al. Exosomes derived from bone-marrow mesenchymal stem cells alleviate cognitive decline in AD-like mice by improving BDNF-related neuropathology. J Neuroinflammation. 2022;19:35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Vatsa P, Negi R, Ansari UA, Khanna VK, Pant AB. Insights of Extracellular vesicles of mesenchymal stem cells: a prospective cell-free Regenerative Medicine for neurodegenerative disorders. Mol Neurobiol. 2022;59:459–74.

    Article  CAS  PubMed  Google Scholar 

  64. Elia CA, Tamborini M, Rasile M, Desiato G, Marchetti S, Swuec P, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wang X, Yang G. Bone marrow mesenchymal stem cells-derived exosomes reduce abeta deposition and improve cognitive function recovery in mice with Alzheimer’s disease by activating sphingosine kinase/sphingosine-1-phosphate signaling pathway. Cell Biol Int. 2021;45:775–84.

    Article  CAS  PubMed  Google Scholar 

  66. Vilaca-Faria H, Marote A, Lages I, Ribeiro C, Mendes-Pinheiro B, Domingues AV, et al. Fractionating stem cells secretome for Parkinson’s disease modeling: is it the whole better than the sum of its parts? Biochimie. 2021;189:87–98.

    Article  CAS  PubMed  Google Scholar 

  67. Sadanandan N, Lee JY, Garbuzova-Davis S. Extracellular vesicle-based therapy for amyotrophic lateral sclerosis. Brain Circ. 2021;7:23–8.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Garbuzova-Davis S, Willing AE, Ehrhart J, Wang L, Sanberg PR, Borlongan CV. Cell-free extracellular vesicles derived from human bone marrow endothelial progenitor cells as potential therapeutics for Microvascular Endothelium Restoration in ALS. Neuromolecular Med. 2020;22:503–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gopinath A, Collins A, Khoshbouei H, Streit WJ. Microglia and other myeloid cells in Central Nervous System Health and Disease. J Pharmacol Exp Ther. 2020;375:154–60.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Nigro A, Colombo F, Casella G, Finardi A, Verderio C, Furlan R. Myeloid extracellular vesicles: messengers from the demented brain. Front Immunol. 2016;7:17.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Williams GP, Schonhoff AM, Jurkuvenaite A, Gallups NJ, Standaert DG, Harms AS. CD4 T cells mediate brain inflammation and neurodegeneration in a mouse model of Parkinson’s disease. Brain. 2021;144:2047–59.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Harms AS, Thome AD, Yan Z, Schonhoff AM, Williams GP, Li X, et al. Peripheral monocyte entry is required for alpha-synuclein induced inflammation and neurodegeneration in a model of Parkinson disease. Exp Neurol. 2018;300:179–87.

    Article  CAS  PubMed  Google Scholar 

  73. Verderio C, Muzio L, Turola E, Bergami A, Novellino L, Ruffini F, et al. Myeloid microvesicles are a marker and therapeutic target for neuroinflammation. Ann Neurol. 2012;72:610–24.

    Article  CAS  PubMed  Google Scholar 

  74. Agosta F, Dalla Libera D, Spinelli EG, Finardi A, Canu E, Bergami A, et al. Myeloid microvesicles in cerebrospinal fluid are associated with myelin damage and neuronal loss in mild cognitive impairment and Alzheimer disease. Ann Neurol. 2014;76:813–25.

    Article  CAS  PubMed  Google Scholar 

  75. Joshi P, Turola E, Ruiz A, Bergami A, Libera DD, Benussi L, et al. Microglia convert aggregated amyloid-beta into neurotoxic forms through the shedding of microvesicles. Cell Death Differ. 2014;21:582–93.

    Article  CAS  PubMed  Google Scholar 

  76. Robling AG, Bonewald LF. The osteocyte: New insights. Annu Rev Physiol. 2020;82:485–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Bonewald LF. The amazing osteocyte. J Bone Min Res. 2011;26:229–38.

    Article  CAS  Google Scholar 

  78. Dallas SL, Prideaux M, Bonewald LF. The osteocyte: an endocrine cell … and more. Endocr Rev. 2013;34:658–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang ZX, Luo ZW, Li FX, Cao J, Rao SS, Liu YW, et al. Aged bone matrix-derived extracellular vesicles as a messenger for calcification paradox. Nat Commun. 2022;13:1453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kyrkanides S, Tallents RH, Miller JN, Olschowka ME, Johnson R, Yang M, et al. Osteoarthritis accelerates and exacerbates Alzheimer’s disease pathology in mice. J Neuroinflammation. 2011;8:112.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Li S, Liu B, Zhang L, Rong L. Amyloid beta peptide is elevated in osteoporotic bone tissues and enhances osteoclast function. Bone. 2014;61:164–75.

    Article  CAS  PubMed  Google Scholar 

  82. Wu XY, Wu XP, Xie H, Zhang H, Peng YQ, Yuan LQ, et al. Age-related changes in biochemical markers of bone turnover and gonadotropin levels and their relationship among Chinese adult women. Osteoporos Int. 2010;21:275–85.

    Article  PubMed  Google Scholar 

  83. Sundaram VK, Schutza V, Schroter NH, Backhaus A, Bilsing A, Joneck L, et al. Adipo-glial signaling mediates metabolic adaptation in peripheral nerve regeneration. Cell Metab. 2023;35:2136–52. e2139.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 2019;15:507–24.

    Article  CAS  PubMed  Google Scholar 

  85. Mazon JN, de Mello AH, Ferreira GK, Rezin GT. The impact of obesity on neurodegenerative diseases. Life Sci. 2017;182:22–8.

    Article  CAS  PubMed  Google Scholar 

  86. Procaccini C, Santopaolo M, Faicchia D, Colamatteo A, Formisano L, de Candia P, et al. Role of metabolism in neurodegenerative disorders. Metabolism. 2016;65:1376–90.

    Article  CAS  PubMed  Google Scholar 

  87. Caron A, Lee S, Elmquist JK, Gautron L. Leptin and brain-adipose crosstalks. Nat Rev Neurosci. 2018;19:153–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Rizzo MR, Fasano R, Paolisso G. Adiponectin and Cognitive decline. Int J Mol Sci. 2020;21:2010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Wardzinski EK, Kistenmacher A, Melchert UH, Jauch-Chara K, Oltmanns KM. Impaired brain energy gain upon a glucose load in obesity. Metabolism. 2018;85:90–6.

    Article  CAS  PubMed  Google Scholar 

  90. Moy GA, McNay EC. Caffeine prevents weight gain and cognitive impairment caused by a high-fat diet while elevating hippocampal BDNF. Physiol Behav. 2013;109:69–74.

    Article  CAS  PubMed  Google Scholar 

  91. Budni J, Bellettini-Santos T, Mina F, Garcez ML, Zugno AI. The involvement of BDNF, NGF and GDNF in aging and Alzheimer’s disease. Aging Dis. 2015;6:331–41.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Guo DH, Yamamoto M, Hernandez CM, Khodadadi H, Baban B, Stranahan AM. Visceral adipose NLRP3 impairs cognition in obesity via IL-1R1 on CX3CR1 + cells. J Clin Invest. 2020;130:1961–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Crewe C, Scherer PE. Intercellular and interorgan crosstalk through adipocyte extracellular vesicles. Rev Endocr Metab Disord. 2022;23:61–9.

    Article  CAS  PubMed  Google Scholar 

  94. Katsuda T, Oki K, Ochiya T. Potential application of extracellular vesicles of human adipose tissue-derived mesenchymal stem cells in Alzheimer’s disease therapeutics. Methods Mol Biol. 2015;1212:171–81.

    Article  PubMed  Google Scholar 

  95. Bi Y, Lin X, Liang H, Yang D, Zhang X, Ke J, et al. Human adipose tissue-derived mesenchymal stem cells in Parkinson’s Disease: inhibition of T Helper 17 cell differentiation and regulation of Immune Balance towards a Regulatory T cell phenotype. Clin Interv Aging. 2020;15:1383–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Garcia-Contreras M, Thakor AS. Human adipose tissue-derived mesenchymal stem cells and their extracellular vesicles modulate lipopolysaccharide activated human microglia. Cell Death Discov. 2021;7:98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Bonafede R, Turano E, Scambi I, Busato A, Bontempi P, Virla F et al. ASC-exosomes ameliorate the disease progression in SOD1(G93A) murine model underlining their potential therapeutic use in human ALS. Int J Mol Sci. 2020;21:3651.

  98. Yildirim S, Oylumlu E, Ozkan A, Sinen O, Bulbul M, Goksu ET, et al. Zinc (zn) and adipose-derived mesenchymal stem cells (AD-MSCs) on MPTP-induced Parkinson’s disease model: a comparative evaluation of behavioral and immunohistochemical results. Neurotoxicology. 2023;97:1–11.

    Article  CAS  PubMed  Google Scholar 

  99. Wang J, Li L, Zhang Z, Zhang X, Zhu Y, Zhang C, et al. Extracellular vesicles mediate the communication of adipose tissue with brain and promote cognitive impairment associated with insulin resistance. Cell Metab. 2022;34:1264–79.

    Article  CAS  PubMed  Google Scholar 

  100. Batabyal RA, Bansal A, Cechinel LR, Authelet K, Goldberg M, Nadler E et al. Adipocyte-derived small extracellular vesicles from patients with Alzheimer disease carry miRNAs predicted to target the CREB signaling pathway in neurons. Int J Mol Sci. 2023;24:14024.

  101. Liu JH, Chen CY, Liu ZZ, Luo ZW, Rao SS, Jin L, et al. Extracellular vesicles from child gut microbiota enter into bone to preserve bone Mass and Strength. Adv Sci (Weinh). 2021;8:2004831.

    Article  CAS  PubMed  Google Scholar 

  102. Tilocca B, Pieroni L, Soggiu A, Britti D, Bonizzi L, Roncada P, et al. Gut-Brain Axis and Neurodegeneration: state-of-the-art of Meta-Omics sciences for Microbiota characterization. Int J Mol Sci. 2020;21:4045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Zhu S, Jiang Y, Xu K, Cui M, Ye W, Zhao G, et al. The progress of gut microbiome research related to brain disorders. J Neuroinflammation. 2020;17:25.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Socala K, Doboszewska U, Szopa A, Serefko A, Wlodarczyk M, Zielinska A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 2021;172:105840.

    Article  CAS  PubMed  Google Scholar 

  105. Tyler Patterson T, Grandhi R. Gut microbiota and neurologic diseases and injuries. Adv Exp Med Biol. 2020;1238:73–91.

    Article  CAS  PubMed  Google Scholar 

  106. Sun M, Ma K, Wen J, Wang G, Zhang C, Li Q, et al. A review of the brain-gut-Microbiome Axis and the potential role of Microbiota in Alzheimer’s Disease. J Alzheimers Dis. 2020;73:849–65.

    Article  PubMed  Google Scholar 

  107. Shen H, Guan Q, Zhang X, Yuan C, Tan Z, Zhai L, et al. New mechanism of neuroinflammation in Alzheimer’s disease: the activation of NLRP3 inflammasome mediated by gut microbiota. Prog Neuropsychopharmacol Biol Psychiatry. 2020;100:109884.

    Article  CAS  PubMed  Google Scholar 

  108. Beydoun MA, Beydoun HA, Elbejjani M, Dore GA, Zonderman AB. Helicobacter pylori seropositivity and its association with incident all-cause and Alzheimer’s disease dementia in large national surveys. Alzheimers Dement. 2018;14:1148–58.

    Article  PubMed  Google Scholar 

  109. Huang HK, Wang JH, Lei WY, Chen CL, Chang CY, Liou LS. Helicobacter pylori infection is associated with an increased risk of Parkinson’s disease: a population-based retrospective cohort study. Parkinsonism Relat Disord. 2018;47:26–31.

    Article  CAS  PubMed  Google Scholar 

  110. Yang X, Yu D, Xue L, Li H, Du J. Probiotics modulate the microbiota-gut-brain axis and improve memory deficits in aged SAMP8 mice. Acta Pharm Sin B. 2020;10:475–87.

    Article  CAS  PubMed  Google Scholar 

  111. Akbari E, Asemi Z, Daneshvar Kakhaki R, Bahmani F, Kouchaki E, Tamtaji OR, et al. Effect of Probiotic supplementation on cognitive function and metabolic status in Alzheimer’s Disease: a Randomized, double-blind and controlled trial. Front Aging Neurosci. 2016;8:256.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Blacher E, Bashiardes S, Shapiro H, Rothschild D, Mor U, Dori-Bachash M, et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature. 2019;572:474–80.

    Article  CAS  PubMed  Google Scholar 

  113. Ou Z, Deng L, Lu Z, Wu F, Liu W, Huang D, et al. Protective effects of Akkermansia muciniphila on cognitive deficits and amyloid pathology in a mouse model of Alzheimer’s disease. Nutr Diabetes. 2020;10:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Gazerani P. Probiotics for Parkinson’s Disease. Int J Mol Sci. 2019;20:4121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Iyaswamy A, Lu K, Guan XJ, Kan Y, Su C, Liu J et al. Impact and advances in the role of bacterial extracellular vesicles in neurodegenerative disease and its therapeutics. Biomedicines. 2023;11:2056.

  116. Sun B, Sawant H, Borthakur A, Bihl JC. Emerging therapeutic role of gut microbial extracellular vesicles in neurological disorders. Front Neurosci. 2023;17:1241418.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Zhao L, Ye Y, Gu L, Jian Z, Stary CM, Xiong X. Extracellular vesicle-derived miRNA as a novel regulatory system for bi-directional communication in gut-brain-microbiota axis. J Transl Med. 2021;19:202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Wei S, Peng W, Mai Y, Li K, Wei W, Hu L, et al. Outer membrane vesicles enhance tau phosphorylation and contribute to cognitive impairment. J Cell Physiol. 2020;235:4843–55.

    Article  CAS  PubMed  Google Scholar 

  119. Xie JH, Cools L, Van Imschoot G, Van Wonterghem E, Pauwels MJ, Vlaeminck I et al. Helicobacter pylori-derived outer membrane vesicles contribute to Alzheimer’s disease pathogenesis via C3-C3aR signalling. J Extracell Vesicles. 2023;12:e12306.

  120. Park AM, Tsunoda I. Helicobacter pylori infection in the stomach induces neuroinflammation: the potential roles of bacterial outer membrane vesicles in an animal model of Alzheimer’s disease. Inflamm Regen. 2022;42:39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lee KE, Kim JK, Han SK, Lee DY, Lee HJ, Yim SV, et al. The extracellular vesicle of gut microbial Paenalcaligenes hominis is a risk factor for vagus nerve-mediated cognitive impairment. Microbiome. 2020;8:107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Cuesta CM, Guerri C, Urena J, Pascual M. Role of Microbiota-Derived Extracellular vesicles in Gut-Brain communication. Int J Mol Sci. 2021;22:4235.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Choi J, Kim YK, Han PL. Extracellular vesicles derived from Lactobacillus plantarum increase BDNF expression in cultured hippocampal neurons and produce antidepressant-like effects in mice. Exp Neurobiol. 2019;28:158–71.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Kwon H, Lee EH, Park SY, Park JY, Hong JH, Kim EK, et al. Lactobacillus-derived extracellular vesicles counteract Abeta42-induced abnormal transcriptional changes through the upregulation of MeCP2 and Sirt1 and improve Abeta pathology in Tg-APP/PS1 mice. Exp Mol Med. 2023;55:2067–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Park SY, Kim DS, Kim HM, Lee JK, Hwang DY, Kim TH, et al. Human mesenchymal stem cell-derived extracellular vesicles promote neural differentiation of neural progenitor cells. Int J Mol Sci. 2022;23:7047.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lai J, Huang C, Guo Y, Rao L. Engineered extracellular vesicles and their mimics in cardiovascular diseases. J Control Release. 2022;347:27–43.

    Article  CAS  PubMed  Google Scholar 

  127. Mager SELA, Breakefield I, Wood XO. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12:347–57.

    Article  PubMed  Google Scholar 

  128. Rehman FU, Liu Y, Zheng M, Shi B. Exosomes based strategies for brain drug delivery. Biomaterials. 2023;293:121949.

    Article  CAS  PubMed  Google Scholar 

  129. Hernando S, Santos-Vizcaino E, Igartua M, Hernandez RM. Targeting the central nervous system: from synthetic nanoparticles to extracellular vesicles-focus on Alzheimer’s and Parkinson’s disease. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2023;15:e1898.

    Article  CAS  PubMed  Google Scholar 

  130. Keaney J, Campbell M. The dynamic blood-brain barrier. FEBS J. 2015;282:4067–79.

    Article  CAS  PubMed  Google Scholar 

  131. Banks WA, Sharma P, Bullock KM, Hansen KM, Ludwig N, Whiteside TL. Transport of extracellular vesicles across the blood-brain barrier: Brain pharmacokinetics and effects of inflammation. Int J Mol Sci. 2020;21:4407.

  132. Yin T, Liu Y, Ji W, Zhuang J, Chen X, Gong B, et al. Engineered mesenchymal stem cell-derived extracellular vesicles: a state-of-the-art multifunctional weapon against Alzheimer’s disease. Theranostics. 2023;13:1264–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Gao W, Li F, Liu L, Xu X, Zhang B, Wu Y, et al. Endothelial colony-forming cell-derived exosomes restore blood-brain barrier continuity in mice subjected to traumatic brain injury. Exp Neurol. 2018;307:99–108.

    Article  CAS  PubMed  Google Scholar 

  134. Li Q, Niu X, Yi Y, Chen Y, Yuan J, Zhang J, et al. Inducible pluripotent stem cell-derived small extracellular vesicles rejuvenate senescent blood-brain barrier to protect against ischemic stroke in aged mice. ACS Nano. 2023;17:775–89.

    Article  CAS  PubMed  Google Scholar 

  135. Li Y, Liu B, Zhao T, Quan X, Han Y, Cheng Y, et al. Comparative study of extracellular vesicles derived from mesenchymal stem cells and brain endothelial cells attenuating blood-brain barrier permeability via regulating Caveolin-1-dependent ZO-1 and Claudin-5 endocytosis in acute ischemic stroke. J Nanobiotechnol. 2023;21:70.

    Article  CAS  Google Scholar 

  136. Hong CG, Chen ML, Duan R, Wang X, Pang ZL, Ge LT, et al. Transplantation of nasal olfactory mucosa mesenchymal stem cells benefits Alzheimer’s Disease. Mol Neurobiol. 2022;59:7323–36.

    Article  CAS  PubMed  Google Scholar 

  137. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–5.

    Article  CAS  PubMed  Google Scholar 

  138. Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Kim JK, Youn YJ, Lee YB, Kim SH, Song DK, Shin M, et al. Extracellular vesicles from dHL-60 cells as delivery vehicles for diverse therapeutics. Sci Rep. 2021;11:8289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Quah BJ, O’Neill HC. The immunogenicity of dendritic cell-derived exosomes. Blood Cells Mol Dis. 2005;35:94–110.

    Article  CAS  PubMed  Google Scholar 

  141. Zhao C, Pan Y, Yu G, Zhao XZ, Chen X, Rao L. Vesicular antibodies: shedding light on antibody therapeutics with cell membrane nanotechnology. Adv Mater. 2023;35:e2207875.

    Article  PubMed  Google Scholar 

  142. Cui GH, Guo HD, Li H, Zhai Y, Gong ZB, Wu J, et al. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Huang T, Jia Z, Fang L, Cheng Z, Qian J, Xiong F, et al. Extracellular vesicle-derived mir-511-3p from hypoxia preconditioned adipose mesenchymal stem cells ameliorates spinal cord injury through the TRAF6/S1P axis. Brain Res Bull. 2022;180:73–85.

    Article  CAS  PubMed  Google Scholar 

  144. Jeske R, Liu C, Duke L, Canonicco Castro ML, Muok L, Arthur P, et al. Upscaling human mesenchymal stromal cell production in a novel vertical-wheel bioreactor enhances extracellular vesicle secretion and cargo profile. Bioact Mater. 2023;25:732–47.

    CAS  PubMed  Google Scholar 

  145. Meng QF, Tai W, Tian M, Zhuang X, Pan Y, Lai J, et al. Inhalation delivery of dexamethasone with iSEND nanoparticles attenuates the COVID-19 cytokine storm in mice and nonhuman primates. Sci Adv. 2023;9:eadg3277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Elsharkasy OM, Nordin JZ, Hagey DW, de Jong OG, Schiffelers RM, Andaloussi SE, et al. Extracellular vesicles as drug delivery systems: why and how? Adv Drug Deliv Rev. 2020;159:332–43.

    Article  CAS  PubMed  Google Scholar 

  147. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16:748–59.

    Article  CAS  PubMed  Google Scholar 

  148. Meng W, He C, Hao Y, Wang L, Li L, Zhu G. Prospects and challenges of extracellular vesicle-based drug delivery system: considering cell source. Drug Deliv. 2020;27:585–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Gorgens A, Corso G, Hagey DW, Jawad Wiklander R, Gustafsson MO, Felldin U, et al. Identification of storage conditions stabilizing extracellular vesicles preparations. J Extracell Vesicles. 2022;11:e12238.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Zhu Y, Huang R, Wang D, Yu L, Liu Y, Huang R, et al. EVs-mediated delivery of CB2 receptor agonist for Alzheimer’s disease therapy. Asian J Pharm Sci. 2023;18:100835.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Qi Y, Guo L, Jiang Y, Shi Y, Sui H, Zhao L. Brain delivery of quercetin-loaded exosomes improved cognitive function in AD mice by inhibiting phosphorylated tau-mediated neurofibrillary tangles. Drug Deliv. 2020;27:745–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Wang H, Sui H, Zheng Y, Jiang Y, Shi Y, Liang J, et al. 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;11:7481–96.

    Article  CAS  PubMed  Google Scholar 

  153. Xu F, Wu Y, Yang Q, Cheng Y, Xu J, Zhang Y, et al. Engineered Extracellular vesicles with SHP2 high expression promote Mitophagy for Alzheimer’s Disease Treatment. Adv Mater. 2022;34:e2207107.

    Article  PubMed  Google Scholar 

  154. Qu M, Lin Q, Huang L, Fu Y, Wang L, He S, et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J Control Release. 2018;287:156–66.

    Article  CAS  PubMed  Google Scholar 

  155. Hampel H, Vassar R, De Strooper B, Hardy J, Willem M, Singh N, et al. The beta-secretase BACE1 in Alzheimer’s Disease. Biol Psychiatry. 2021;89:745–56.

    Article  CAS  PubMed  Google Scholar 

  156. Jahangard Y, Monfared H, Moradi A, Zare M, Mirnajafi-Zadeh J, Mowla SJ. Therapeutic effects of transplanted exosomes containing miR-29b to a rat model of Alzheimer’s Disease. Front Neurosci. 2020;14:564.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Zhai L, Shen H, Sheng Y, Guan Q. ADMSC Exo-MicroRNA-22 improve neurological function and neuroinflammation in mice with Alzheimer’s disease. J Cell Mol Med. 2021;25:7513–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Geng Y, Long X, Zhang Y, Wang Y, You G, Guo W, et al. FTO-targeted siRNA delivery by MSC-derived exosomes synergistically alleviates dopaminergic neuronal death in Parkinson’s disease via m6A-dependent regulation of ATM mRNA. J Transl Med. 2023;21:652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Xiao Y, Tang Z, Huang X, Chen W, Zhou J, Liu H, et al. Emerging mRNA technologies: delivery strategies and biomedical applications. Chem Soc Rev. 2022;51:3828–45.

    Article  CAS  PubMed  Google Scholar 

  160. Izadpanah M, Dargahi L, Ai J, Asgari Taei A, Ebrahimi Barough S, Mowla SJ, et al. Extracellular vesicles as a Neprilysin Delivery System Memory Improvement in Alzheimer’s Disease. Iran J Pharm Res. 2020;19:45–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  161. de Godoy MA, Saraiva LM, de Carvalho LRP, Vasconcelos-Dos-Santos A, Beiral HJV, Ramos AB, 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;293:1957–75.

    Article  PubMed  Google Scholar 

  162. Zhao Y, Haney MJ, Gupta R, Bohnsack JP, He Z, Kabanov AV, et al. GDNF-transfected macrophages produce potent neuroprotective effects in Parkinson’s disease mouse model. PLoS ONE. 2014;9:e106867.

    Article  PubMed  PubMed Central  Google Scholar 

  163. He S, Wang Q, Chen L, He YJ, Wang X, Qu S. miR-100a-5p-enriched exosomes derived from mesenchymal stem cells enhance the anti-oxidant effect in a Parkinson’s disease model via regulation of Nox4/ROS/Nrf2 signaling. J Transl Med. 2023;21:747.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Morrell AE, Brown GN, Robinson ST, Sattler RL, Baik AD, Zhen G, et al. Mechanically induced ca(2+) oscillations in osteocytes release extracellular vesicles and enhance bone formation. Bone Res. 2018;6:6.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Gerosa L, Lombardi G. Bone-to-Brain: a round trip in the adaptation to mechanical stimuli. Front Physiol. 2021;12:623893.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (82301625 to X.X. L.; 82272562 to Z.X. W.; 82125023, 82072504 to H. X.; U22A20300, 81971029 to L. S.), the National Key R&D Program of China (2020YFC2008500 to L. S.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, X.X. L., Z.X. W., and H. X.; writing—original draft preparation, X.X. L.; writing—review and editing, Z.X. W., H. X., and L. S.; figures and tables, X.X. L., and M.D. W; and funding acquisition, X.X. L., Z.X. W., H. X., and L. S. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Hui Xie or Zhenxing Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X., Shen, L., Wan, M. et al. Peripheral extracellular vesicles in neurodegeneration: pathogenic influencers and therapeutic vehicles. J Nanobiotechnol 22, 170 (2024). https://doi.org/10.1186/s12951-024-02428-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02428-1

Keywords