Back to Journals » Journal of Inflammation Research » Volume 15

The Emerging Role of the Interaction of Extracellular Vesicle and Autophagy—Novel Insights into Neurological Disorders

Authors Wei W , Pan Y , Yang X, Chen Z, Heng Y, Yang B, Pu M, Zuo J, Lai Z, Tang Y, Xin W 

Received 17 February 2022

Accepted for publication 1 June 2022

Published 9 June 2022 Volume 2022:15 Pages 3395—3407

DOI https://doi.org/10.2147/JIR.S362865

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Adam D Bachstetter



Wei Wei,1,2,* Yongli Pan,2,3,* Xinyu Yang,4 Zhonglun Chen,1 Yue Heng,1 Bufan Yang,1 Mingjun Pu,1 Jiacai Zuo,1 Zhuhong Lai,5 Yufeng Tang,1 Wenqiang Xin2,4

1Department of Neurology, Mianyang Central Hospital, Mianyang, Sichuan, People’s Republic of China; 2Department of Neurology, University Medical Center of Göttingen, Georg-August-University of Göttingen, Göttingen, Lower Saxony, Germany; 3Department of Neurology, Weifang Medical University, Weifang, Shandong, People’s Republic of China; 4Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China; 5Department of Cardiology, Mianyang Central Hospital, Mianyang, Sichuan, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Wenqiang Xin, Department of Neurosurgery, Tianjin Medical University General Hospital, Anshan Road No. 154, Tianjin, 300052, People’s Republic of China, Tel +86– 18526201182, Fax +86– 2260362062, Email [email protected] Wei Wei, Department of Neurology, Mianyang Central Hospital, Changjia Lane No. 12, Mianyang, 621000, People’s Republic of China, Tel +86– 15983634286, Fax +86-8162222566, Email [email protected]

Abstract: Eukaryotic cells release different types of extracellular vesicles (EVs), including exosomes, apoptotic bodies and microvesicles. EVs carry proteins, lipids and nucleic acids specific to cells and cell states. Autophagy is an intracellular degradation process, which, along with EVs, can significantly affect the development and progression of neurological diseases and, therefore, has been the hotspot. Generally, EVs and autophagy are closely associated. EVs and autophagy can interact with each other. On the one hand, the level of autophagy in target cells is closely related to the secretion and transport of EVs. In another, the application of EVs provides a great opportunity for adjuvant treatment of neurological disorders, for which autophagy is an excellent target. EVs can release their cargos into target cells, which, in turn, regulate the autophagic level of target cells through autophagy-related proteins directly and the non-coding RNA, signal transducer and activator of transcription 3 (STAT3), phosphodiesterase enzyme (PDE) 1-B, etc. signaling pathways indirectly, thus regulating the development of related neurological disorders.

Keywords: extracellular vesicle, autophagy, neurological disorders, non-coding RNA, STAT3, PDE1-B

Introduction

Extracellular vesicles (EVs) are lipid-binding vesicles secreted by cells into the extracellular space, mainly composed of microvesicles (MVs), exosomes and apoptotic bodies.1,2 They transport the cargos of proteins, nucleic acids, lipids, metabolites and organelles from the parent cell, and take an important role in biological functions.3,4 EVs are released by nearly all cell types, such as oligodendrocytes, neurons, astrocytes, microglia and endothelial cells in the central nervous system (CNS).5–7 EVs can stimulate neuronal development by facilitating neuron-glia contact and take a role in the pathophysiology of several neurological diseases, including stroke, multiple sclerosis (MS), Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), traumatic brain injury (TBI) and prion diseases.8–11 Autophagy is a self-degradation process, which is a dynamic process that plays a key role in cell homeostasis by adapting cell metabolism to a stressful environment.12 There are different types of autophagy, including typical macroautophagy, chaperone-mediated autophagy, and less studied microautophagy.13 Meanwhile, autophagy is also associated with other inner membrane systems and signaling pathways to regulate endocytosis, exocytosis, and even the hydrolysis of biomolecules.14 Growing evidence indicated that EVs participate in the excretion, degradation, and recovery of biomolecules, and play an important role in autophagy to promote cell survival.15,16 Recent studies have reported the ability of EVs to cooperate with autophagy flux for maintaining cell homeostasis. For example, adipose-derived mesenchymal stem cells (ADMSCs) or ADMSC-secreted EVs could induce neuroprotection by regulating autophagic flux through secreting EVs containing microRNA (miR) in ischemic stroke.8 The present state of research into the involvement between EVs and autophagy in neurological disorders is summarized in this section, and describes the signaling pathways involved in mediating EVs-modulated autophagy.

EVs and Neurological Disorders

The Brief Course on Biogenesis of EVs

EVs, which serve as membrane-enclosed nanoscale particles, carry a variety of cargo, such as peptides, lipids, proteins nucleic acids (DNA, mRNA, short non-coding and long non-coding RNAs).17 They act as regulation factors by their phenotypic effects on recipient cells and deliver information between cells both in a paracrine or endocrine manner.18 These small bioactive molecules can also bear biomolecules of their parent cell of origin and may provide diagnostic and prognostic value in CNS diseases.19,20 Multiple types of EVs exist, which can be categorized into 3 different types, the basis on their biogenesis, size, and isolation method: exosomes, microvesicles, and apoptotic bodies.21 The exosomes with heterogeneous sizes (30–150nm) and compositions that elicit differential molecular and biological properties are generated from multivesicular bodies (MVB) in the endosomal system, which bud directly from the cell membrane.22 Regarding the microvesicles (50–1000nm), they are generated by an outward budding from the plasma membrane of the cell and occur selectively in the lipid-rich microdomains of the membrane.21 Apoptotic bodies (50–5000nm) are another type of EVs that are larger than exosomes and microvesicles, which are exclusively released from the cells undergoing apoptotic cell clearance during the last steps of the apoptosis process including cell dismantling and recycling of biomolecule building blocks.23,24

The Vital Role of EVs in Neurological Disorders

Studies on EVs have contributed to broadening our current acknowledge of the physiology and pathology of nervous system disease. Depending on a cell of origin, microenvironment and the status of the disease, EVs can present a variety of functions. EVs, derived from neurons, astrocytes, microglia and oligodendrocytes, serve as regulators and mediate cell to cell communication.25 Under neurophysiological conditions, neuronal EVs can regulate the differentiation of neural axons by transferring neuron-specific cargoes to a variety of glial cells and regulating relevant functions.26 For instance, miR-124-3p can be delivered by neuronal EVs from primary neurons to astrocytes and increase the expression of the glutamate transporters.27 Moreover, EVs secreted by microglia can transfer nervous growth factors to neurons.28 Microglia also absorbs myelin debris through oligodendrocytes-derived EVs.29 Nevertheless, EVs show several different properties under pathological conditions. On the one hand, EVs exert protective effects on neurons and remove pathological deposition proteins. For example, the relationship between alternative EVs molecules and prognosis of stroke has already been explored.30 This characteristic can be utilized to distinguish several substyles of stroke with high accuracies, such as spontaneous intraparenchymal hemorrhage, aneurysmal subarachnoid hemorrhage and ischemia stroke.31 Additionally, EVs have great potential in the treatment of stroke, compared to the narrow time window of the traditional method. Neural progenitor cell (NPCs)-EVs, as a critical factor, participate in neurogenesis and neural restoration in post-stroke.32 Furthermore, the effect of NSC-EVs has also been evaluated in an animal ischemia stroke model.33 The result of pathologic and symptomatic status improved significantly after administrating EVs after 24 hours, including alleviating lesion volume and brain swelling. Likewise, Xin et al have demonstrated that gene-modified EVs can enhance neuroplasticity and function recovery after MCAO treatment in rat.34 On the other hand, EVs can accelerate the pathological process of neurological disorders including inflammation, apoptosis, and autophagy. Microglia transfers EVs to astrocytes during neuroinflammation, which initiates the trigger to activate A1 astrocytes, leading to neuronal damage.35 Besides, ceramide-containing EVs from astrocytes can cause neural apoptosis during AD pathological proteins.36

Autophagy and Neurological Disorders

The Brief Course on Autophagy

Autophagy is a general term that mainly refers to self-eating cellular catabolism,37 which is a process induced by changes in the internal conditions of cells,38 such as starvation, hypoxia nutrient deficiencies, and infection,39 resulting in degrade toxic proteins, damaged organelles and invading pathogens via the lysosomal pathway.40 It plays a vital role in stabilizing cell homeostasis by orderly degradation and recycling cellular components. As much as we know, three different kinds of autophagy are found in mammalian cells according to their methods of substrate delivery: macroautophagy, microautophagy and chaperone-mediated autophagy (CMA).41 Macroautophagy is an intracellular degradation system for the majority of proteins and some organelles,42 which means it can break down cargo and then allow for the recycling of the resulting macromolecules, and the recycling of substrates through macroautophagy has an important role in maintaining cellular homeostasis.43 Microautophagy, in which the lysosomal degradative process uses autophagic tubes to mediate both invagination and vesicle scission into the lumen by direct engulfment of cytoplasmic cargo.44 It also exerts an effect on cellular homeostasis. The major role of CMA in cellular metabolism is supposed to supply amino acids after protein degradation. However, some recent researchers find that the energy metabolism can also be significantly altered after the function of CMA impairment.45

A Double-Edged Sword Role of Autophagy and Possible Signaling Pathway

In mammals, cells in the CNS are especially vulnerable to damage. Autophagy is the main response against aggregate-prone proteins, defective organelles and insoluble protein aggregates, which plays an important housekeeping role in the CNS.46 Despite it has been unclear whether autophagy plays a potential beneficial or harmful role in the CNS, there is no doubt that autophagy critically contributes to neuronal fate.47 Under the ischemic condition, neurons are particularly vulnerable and autophagy is thought to be activated by the clearance of protein aggregates and damaged mitochondria and preservation of energy balance.48 Meanwhile, numerous direct evidence has been verified that autophagy could be a therapeutic target in ischemic stroke. Researchers reported that rapamycin stimulates autophagy, which then improves mitochondrial function and reduces infarct volume, brain edema, and motor impairments resulting from ischemia.49,50 However, this function could be reversed by administrating 3-methyl-adenine (3-MA) through intracerebroventricular injection, an autophagy inhibitor.51 Some other studies find that autophagy is also related to the promotion of cell death. It is possible that excessive up-regulation of autophagy and long-term autophagy eventually result in self-digestion or have harmful effects.52 Interestingly, in some AD and PD patients, accumulation of autophagy vesicles (autophagosomes and lysosomes) has been observed, leaving the question of whether the suppressive or excessive autophagy leads to the accumulation of vesicles.53,54 Multiple signaling pathways can be activated under the stressed condition that subsequently feeds into the autophagy pathway47 mainly in neurodegenerative diseases. ULK1/2 forms a complex with ATG13, ATG101, and focal adhesion kinase family-interacting protein of 200 kDa (FIP200). Mammalian target of rapamycin complex 1 (mTORC1) and AMP-dependent protein kinase (AMPK) can be activated by nutrients, growth factors, and AMP/ATP respectively, which, regulate the ULK1/2 complex through a series of phosphorylation events,55 thereby in response to the autophagy activation. Some studies suggest that the PI3K/Akt/mTOR pathway could regulate ischemia injury.56,57 One research also has elaborated that knockout of p50 (NF-κB1) enhanced autophagy by repression of mTOR in cerebral ischemic mice58 and hypoxia-inducible factor 1 (HIF-1), reactive oxygen species (ROS), and AMP-activated protein kinase (AMPK) are all involved in response to hypoxia during cerebral ischemia.59,60 One analysis of brain tissue from AD patients found the role of Keap1 in autophagosomes.61 Besides, in HD models, overexpression of IRE1 kinase leads to impaired autophagic flux62 and IRE1 can recruit TRAF2/ASK same as activation of JNK, disrupting its interaction with BECN1, therefore activating autophagy.63

The Crosstalk of Impact of Autophagy on EVs

Cells use EVs to communicate with the environment and surrounding cells by carrying information such as lipids, proteins, or nucleic acids.64,65 Autophagy plays a vital role in the synthesis and degradation of EVs. The mechanism of EVs secretion and transport seems to be closely related to the formation of autophagosomes.64 It has been reported that extracellular components can be degraded through the endo/exosome pathway, while intracellular components can be delivered to lysosomes through the process of autophagy, ultimately leading to the degradation of lysosomal contents.66 In addition, autophagosomes not only have a strong ability to fuse with lysosomes but also can fuse with multivesicular bodies to form amphiphiles, which eventually fuse with lysosomes and dissolve the inner material of intraluminal vesicles (ILVs).64 Therefore, intracellular autophagy plays a decisive role in the secretion and transport of EVs. Elevated levels of autophagy significantly inhibited EVs release due to increased fusion of multivesicular bodies with autophagic vacuoles.67 In contrast to the release of EVs, the precursor MVBs of exosomes can be cleared through the autophagy-lysosome pathway by direct fusion with lysosomes or autophagosomes. This is because multivesicular bodies directly enter the autophagy pathway after autophagy activation, and then significantly reduce the release of EVs68(Figure 1).

Figure 1 EVs biogenesis and autophagy in neurological diseases. Existing studies have shown that abnormal protein accumulation and aggregation are hallmarks of various neurological diseases, while exosome release and autophagic degradation are two ways to clear them, and there are multiple possible interactions between autophagy and exosome biogenesis: (I) Macroautophagy begins with phagophore formation and expansion: phagocytosis of cytoplasmic proteins and organelles when VPS34, beclin-1 and ATG14 form a complex and initiate phagophore nucleation and formation. Then, with the assistance of ATG12, Rab11 and LC3 proteins, it leads to the formation of autophagosomes. The autophagosome moves along microtubules, during which it can fuse with MVBs and exchange substances to form two bodies. Both autolysate and amphibian formation are controlled by ESCRT proteins. It then fuses with the lysosome to degrade the engulfed contents.120 (II) Maturation of early endosomes produces MVBs, late endocytic compartments containing numerous ILVs. Fusion of MVBs to the plasma membrane results in the release of ILVs into the extracellular space as exosomes. (III) Amphisomes can fuse with the plasma membrane and secrete their contents. Shown is the autophagy-dependent secretion of ANXA2, where the amphisome intermediate is required for ANXA2 release in exosomes. This image is adapted from previous studies64,120,121 published under the Creative Common Attribution License.

Abbreviations: ANXA2, annexin A2; ESCRT, endosomal sorting complexes required for transport proteins; MVBs, multivesicular bodies; ILVs, intraluminal vesicles.

In neurodegenerative diseases, the aberrant protein aggregation of α-synuclein is thought to be critical.69 EVs are considered to serve as a vehicle for this abnormal protein to transport from cell to cell. The current studies showed that EVs with α-synuclein are fully capable of inducing abnormal protein aggregation in recipient neurons, impairing autophagic flux by upregulating PELI1, which in turn leads to the degradation of LAMP-2.69–71 Willén et al indicated that AD-linked β-amyloid (Aβ) causes EV enlargement and that amyloid fibrils can act in the endocytic pathway of neurons. In turn, altering EVs can also lead to the accumulation and aggregation of Aβ.72 In addition, the EVs containing α-synuclein and beta-amyloid, as well as other pathogenic proteins, are involved in mediating autophagy and inflammation, and a similar discovery of their potential role is also confirmed in AD.73–77 Houtman et al77 and Ahmed et al76 found that the inflammatory response promoted by the NLRP3-Caspase-1 inflammasome pathway triggers autophagy dysfunction and Aβ accumulation, and can be amplified and regulated by LC3-positive vesicles, further confirming the important role of autophagy in AD pathological progression. Furthermore, recent evidence suggests that aggregation of α-synuclein and the resulting cytotoxicity are hallmarks of PD. The secretory pathway of α-synuclein oligomers is strongly influenced by autophagic activity. Compared with free α-synuclein oligomers, exosome-associated α-synuclein oligomers are more readily taken up by recipient cells and can induce more toxicity. Therefore, preventing α-synuclein exosomes release and regulating autophagy may be a novel approach to preventing disease transmission in PD.69,78,79 More importantly, by purifying microglia/macrophage EVs from the cerebrospinal fluid of PD patients, they confirmed the presence of α-synuclein oligomers in CD11b+ exosomes and that such exosomes inhibit autophagy by impairing autophagic flux, thereby disrupting intracellular homeostasis and exacerbating neurotoxicity, further supporting the involvement of exosomes in α-synuclein-related pathological processes.80 Taken together, the level of autophagy in target cells is closely related to the secretion and transport of EVs.

EVs are Involved in Regulating Autophagy in Neurological Disorders

Stroke

It is known that EVs play multiple roles in ischemic stroke, including inhibiting neuronal apoptosis, regulating autophagy, and reducing inflammation.9,34,81–84 Especially, recent researches show that EVs and EV-cargos can be used as biomarkers for the different ischemic stroke stages and inhibit abnormal autophagy to improve neuroregeneration in preclinical ischemic stroke model.34,85–87 Hence, we summarized EVs secreted from several kinds of cells, such as astrocytes, adipose-derived stem cells (ADSC), MSC and microglia, can alleviate neurological deficits by regulating autophagy in ischemic models (Table 1). Most studies showed theses effects of EVs by using a rat or mouse model in their researches. Pei et al reported that EVs derived from astrocytes could enhance neuron viability, and inhibit OGD-induced apoptosis and levels of TNF-α, IL-6, and IL-1β via regulating autophagy.88 In addition, inhibition or knockdown of PDE1-B significantly enhanced the autophagic flux in BV2 cells, promoted M2, and inhibited the M1 phenotype.89 These EVs derived from conditioned microglia are expected to regulate cortical neuronal survival under ischemic conditions. On the other hand, many EV-miRNAs that improve ischemia-induced neuronal damage via regulating autophagy have been found. Chen et al demonstrated exosomes secreted from ischemic-preconditioned astrocyte (IPAS-EXOs) cultured with neurons exert neuroprotection.90 They observed that these effects depend on circSHOC2 in IPAS-EXOs, suppressed neuronal apoptosis, and ameliorated neuronal damage by regulating autophagy and acting on the miR-7670-3p/SIRT1 axis in vivo model.90 In addition, other researchers also indicated that EVs derived from astrocytes transferred miR-190b or miR-361 could inhibit OGD-induced autophagy and neuronal damage by targeting Atg7 or downregulating the AMPK/mTOR pathway, respectively.91,92 Besides, EVs secreted from ADSCs have been widely researched and shown to exert beneficial effects in reducing infarct size by an intravenous injection.8,9 Kuang et al indicated that miR-25-3p is the highest expressed miRNA in ADMSC-EVs that interacts with the p53 pathway, and miR-25-3p oligonucleotide mimics reduce cell death while anti-oligonucleotides regulate P53-BNIP3 signaling in primary neurons to increase autophagy flux and cell death in cerebral ischemia. Hence, AD-MSCs induce neuroprotection by improved autophagic flux through secreted EVs containing miR-25-3p.8 In addition, miR-30d-5p-enhanced EVs derived from ADSCs inhibit autophagy-mediated microglia polarization to M1, promote M2 microglia/macrophage polarization and reducing infarct size and brain damage.9 Finally, EVs derived from humans are also effective for ischemic impairment. miR-21-3p is significantly down-regulated in human umbilical vein endothelial cell (HUVECs)-EVs during hypoxia/reoxygenation (H/R). The miR-21-3p knockdown can activate autophagy and inhibit cell apoptosis, showing a protective effect on neuron cells treated by H/R.93 Xia et al also illustrated that EVs secreted by MSCs inhibited autophagy and promoted angiogenesis by regulating and activating signal transducer and activator of transcription 3 (STAT3), reducing the infarct size, enhancing angiogenesis and reducing long-term neurological deficits.85

Table 1 Preclinical Studies Assessing the Effect of EVs on the Activation of Autophagy in Stroke

Neurodegenerative Diseases

Current research has demonstrated that EVs are related to disease progression in neurodegenerative diseases. These EVs may have multiple and important physiological functions in AD and PD, from deregulating synaptic activity, promoting demyelination, modulating autophagy impairment and regulating microglia activity.94,95 Recently, research into the effects of EVs on autophagy and the therapy of AD and PD has also been extensively studied (Table 2). EVs isolated from umbilical cord MSCs conditioned medium (ucMSCs-CM) exposed to BV2 microglial cells, showed that ucMSCs-EVs promoted the phagocytosis of BV2 cells, affected the level of autophagy-related proteins (LC3, Beclin-1 and p62) and finally inhibited the proliferation and decreased cell death of BV2 cells induced by Aβ25–35.96 Furthermore, injection of EVs derived from hypoxic MSCs could decrease the activation of astrocytes and microglia, down-regulate proinflammatory cytokines (TNF-α and IL-1β), and up-regulate anti-inflammatory cytokines (IL-4 and IL-10) via regulating the activation of signal transducer and activator of transcription 3 (STAT3) and NF-kB.97 On the other hand, a study uncovered that α-synuclein-containing EVs released by microglia were able to induce protein aggregation in recipient neurons, whereas depleting microglia dramatically suppressed the transmission of α-synuclein in PD. Moreover, α-synuclein also impaired autophagy flux by upregulating PELI1, resulting in degradation of LAMP2 in activated microglia.80 These results suggest that regulation of autophagy, blocking microglial delivery of α-synuclein via exosomal pathways, has the potential to serve as a therapeutic target for PD.98 Zhou et al indicated that enhancing the expression of miR-19a-3p in exosomes derived from SH-SY5Y cells inhibited the autophagy of recipient microglia through the AKT/mTOR signaling pathway.99 Furthermore, using miR-188-3p-enriched EVs derived from ADSC could suppress autophagy and Pyroptosis, whereas increased proliferation via targeting CDK5 and NLRP3 in mice and MN9D cells.100 Other studies have also revealed that α-synuclein secretion in EVs can be affected by regulating autophagy.70,101–103 Finally, substances such as rapamycin, curcumin and manganese can also affect the release of EVs from dopaminergic neurons by regulating autophagy, thereby reducing the aggregation of α-synuclein.71,101,104

Table 2 Preclinical Studies Assessing the Effect of EVs on the Activation of Autophagy in Neurodegenerative Diseases

TBI

There is growing evidence that EVs and autophagy play important roles in TBI, therefore, abnormal EVs secretion and autophagy may lead to further neuronal damage over time.105–107 Recent studies have shown that the autophagy pathway is continuously activated after TBI, which may lead to aggravated neural damage (Table 3). For example, the level of miR-21-5p in neuronal exosomes increased from the acute phase of TBI. The use of this miR-21-5p-enriched neuronal exosomes could inhibit neuronal autophagy activity by inhibiting neuronal autophagy targeting Rab11a, thereby attenuating trauma-induced autophagy-mediated neural damage in vitro.105 Similarly, the increase of miR-124-3p in microglial exosomes after TBI might inhibit neuronal autophagy and prevent neuronal damage by translocating it into neurons.106 Overall, treatment with these miR-enriched EVs may represent a new therapeutic strategy for the treatment of nerve damage after TBI. A schematic illustration of EVs derived from different cell sources and their respective roles in neurological diseases is shown in Figure 2.

Table 3 Preclinical Studies Assessing the Effect of EVs on the Activation of Autophagy in Traumatic Brain Injury

Figure 2 Schematic illustration of EVs derived from different cell sources and their respective roles in neurological diseases. In the central and peripheral system, different donor cells including neurons, microglia, and astrocyte can modulate their respective recipient cells by transferring various extracellular vesicle-cargos through modulating autophagy, thus regulating central nervous system diseases progression and recover.

Abbreviations: ADSCs, adipose-derived stem cells; MSCs, mesenchymal stem cells; EVs, extracellular vesicles; AD, Alzheimer’s disease; TBI, traumatic brain injury.

The Mechanism of EVs in the Effect of Regulating Autophagy

ncRNAs in EVs are Key Players in Regulating Autophagy

Non-coding RNAs (ncRNAs), such as miRs, long non-coding RNAs and circular RNAs (circRNAs), are a large class of RNA transcripts that are transcribed from the genome, however, lack the function of encoding proteins.108 At the RNA level, they can conduct their respective biological effects and play a vital role in cell growth, differentiation, replication, and apoptosis.108,109 ncRNAs are also uncovered in the extracellular milieu, plenty of ncRNAs selectively sorted into EVs potentially regulate specific aspects of autophagy, thus protecting from degradation. Current shreds of evidence have demonstrated that ncRNAs are effective treatment candidates owing to their capacity of promoting neuronal recovery. Studies have demonstrated the effects of autophagy regulation of specific ncRNAs, especially for miRs, which are highly expressed in EVs.110 miRs, the biggest family of ncRNAs containing 20–25 nucleotides, play key roles in the remodeling process under neurological disorders.110 It is reported that miR-21-3p can attenuate brain injuries via multiple mechanisms.111,112 Interestingly, the miR-21-3p expression was downregulated in HUVECs after a hypoxic condition, suggesting that miR-21-3P might be a potential effector.93 To explore its involvement in autophagy pathways played by miR-21-3p in the protective effect of HUVECs-exosomes on hypoxia-treated neural cells, the pathway mediating the effect of miR-21-3p was explored by focusing on the activity of ATG12 protein. Jiang et al conducted a dual-luciferase assay and revealed that miR-21-3p can directly bind to the 3ʹUTR sequence of the ATG12 gene and suppresses ATG12 signaling.93 Likewise, Li et al indicated that the expression of miR-21-5p was increased in EVs derived from HT-22 neurons extracted from the TBI mice brain. EV-miR-21-5p produced a protective effect by suppressing autophagy in HT-22 neurons after scratch injury via directly targeting the Rab11a 3ʹUTR region to inhibit Rab11a-mediated neuronal autophagy.105 Besides that, multiple miRNAs, such as miR124-3p,106 miR19a-3p,99 miR188-3p,100 miR-30d-5p,9 miR-190b,91 miR-7670,90 miR-258 and miR-135a,113 shuttered in respective EVs are involved in regulating autophagy various cell as shown in Tables. Hence, a plethora of miRNAs are found in EVs of which the precise signaling cascades that are regulated are not fully known, yet.

mRNA, Such as STAT3 and PDE1-B, Play an Important Role in the Effect Regulating Autophagy

Phosphodiesterase enzyme (PDE) 1-B is the best characterized among 11 groups of the superfamily of PDE enzymes in mammals.114 PDE, a calcium and calmodulin-dependent phosphodiesterase, limits the intracellular levels of cyclic nucleotides by catalyzing the hydrolysis of cAMP and cGMP. Recently, it has been revealed that PDE1-B is involved in the regulation of autophagy and exosome release.89 Zang et al89 demonstrated a novel mechanism by which vinpocetine, an inhibitor of PDE1-B, regulated microglia-neuron communication via altering autophagy in BV2 cells. Firstly, the PDE1-B expression in microglia was progressively elevated in the peri-infarct region after MCAO. Using vinpocetine can inhibit BV2 microglia M1, promote M2 phenotype, and enhance autophagy in OGD-conditioned BV2 cells, which is associated with the release of exosomes that protects neurons against OGD-induced damage. With regard to signal transducer and activator of transcription 3 (STAT3), located on chromosome 17q21, which was first reported as a transcriptional enhancer of acute-phase genes activated by interleukin 6.115 STAT3, the only embryonic lethal family member of the STAT family, is a prominent nuclear transcription factor that regulates more than 1000 gene expressions.116 STAT3 protein becomes transcriptionally activated primarily by tyrosine phosphorylation, in turn, translocates to the nucleus and targets sequence-specific DNA elements for consequent transcription of target genes.115,116 A great number of studies have endorsed the growing evidence of an important role of STAT3 in autophagy regulation.117–119 Interestingly, the current study has demonstrated that EVs can regulate autophagy as the upstream of STAT3. For example, Xia et al85 indicated that the expression of the LC3-II/LC3-I and Beclin-1 was significantly increased while the P62 protein level was decreased after the stroke model. However, MSCs-EV markedly reversed the levels of autophagy-associated protein levels induced by MCAO. Besides that, the specific autophagosomes characterized by double-membrane structure were increased after MCAO. Notably, the EV group found fewer autophagosomes by TEM. Furthermore, EVs can significantly activate STAT3 after stroke. Xia et al85 used static, a STAT3 inhibitor, to further confirm the role of STAT3 in the preventative effects of EVs on stroke-induced autophagy, and revealed suppression of STAT3 can abolish EV-induced inhibition of autophagy in vivo and in vitro. MSCs-EVs might therefore contribute to inhibiting autophagy by activating STAT3. The ability of EVs from other cells to suppress autophagy, information regarding this aspect, however, is scarce and appears to be limited, for which additional and reliable data is urgently necessary.

Conclusions and Prospects

Generally, EVs and autophagy are closely associated. Cells use EVs to communicate with the environment and surrounding cells by carrying information such as lipids, proteins, or nucleic acids. Autophagy plays a vital role in the synthesis and degradation of EVs, which is closely related to the formation of autophagosomes. In another, the application of EVs provides a great opportunity for adjuvant treatment of neurological disorders, for which autophagy is an excellent target. Although EVs exist in extracellular fluid, they can release their cargos into target cells via endocytosis, receptor-ligand binding, and membrane fusion, which, in turn, regulate the autophagic level of target cells through autophagy-related proteins directly and the ncRNA, STAT3, PDE1-B, etc. signaling pathways indirectly, thus regulating the development of related neurological disorders.

Acknowledgments

We thank the support of the China Scholarship Council for YL Pan, W Wei and WQ Xin.

Disclosure

Wei Wei and Yongli Pan should be considered co-first authors.The authors declare that they have no competing interests.

References

1. Zaborowski MP, Balaj L, Breakefield XO, et al. Extracellular vesicles: composition, biological relevance, and methods of study. Bioscience. 2015;65(8):783–797.

2. Yáñez-Mó M, Siljander PR-M, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4(1):27066. doi:10.3402/jev.v4.27066

3. Van Niel G, d’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228. doi:10.1038/nrm.2017.125

4. Subedi P, Schneider M, Philipp J, et al. Comparison of methods to isolate proteins from extracellular vesicles for mass spectrometry-based proteomic analyses. Anal Biochem. 2019;584:113390. doi:10.1016/j.ab.2019.113390

5. Sharma P, Schiapparelli L, Cline HT. Exosomes function in cell-cell communication during brain circuit development. Curr Opin Neurobiol. 2013;23(6):997–1004. doi:10.1016/j.conb.2013.08.005

6. Fauré J, Lachenal G, Court M, et al. Exosomes are released by cultured cortical neurones. Mol Cell Neurosci. 2006;31(4):642–648. doi:10.1016/j.mcn.2005.12.003

7. Janas AM, Sapoń K, Janas T, et al. Exosomes and other extracellular vesicles in neural cells and neurodegenerative diseases. Biochim Biophys Acta. 2016;1858(6):1139–1151. doi:10.1016/j.bbamem.2016.02.011

8. Kuang Y, Zheng X, Zhang L, et al. Adipose-derived mesenchymal stem cells reduce autophagy in stroke mice by extracellular vesicle transfer of miR-25. J Extracell Vesicles. 2020;10(1):e12024. doi:10.1002/jev2.12024

9. Jiang M, Wang H, Jin M, et al. Exosomes from MiR-30d-5p-ADSCs reverse acute ischemic stroke-induced, autophagy-mediated brain injury by promoting M2 microglial/macrophage polarization. Cell Physiol Biochem. 2018;47(2):864–878.

10. Thompson AG, Gray E, Heman-Ackah SM, et al. Extracellular vesicles in neurodegenerative disease - pathogenesis to biomarkers. Nat Rev Neurol. 2016;12(6):346–357. doi:10.1038/nrneurol.2016.68

11. Lee S, Mankhong S, Kang JH. Extracellular vesicle as a source of Alzheimer’s biomarkers: opportunities and challenges. Int J Mol Sci. 2019;20(7):1728.

12. Kocaturk NM, Gozuacik D. Crosstalk between mammalian autophagy and the ubiquitin-proteasome system. Front Cell Dev Biol. 2018;6:128.

13. Galluzzi L, Baehrecke EH, Ballabio A, et al. Molecular definitions of autophagy and related processes. EMBO Jl. 2017;36(13):1811–1836. doi:10.15252/embj.201796697

14. Mizushima N, Komatsu MJC. Autophagy: renovation of cells and tissues. Cell. 2011;147(4):728–741. doi:10.1016/j.cell.2011.10.026

15. Ponpuak M, Mandell MA, Kimura T, et al. Secretory autophagy. Curr Opin in Cell Biol. 2015;35:106–116. doi:10.1016/j.ceb.2015.04.016

16. Lehmann BD, Paine MS, Brooks AM, et al. Senescence-associated exosome release from human prostate cancer cells. Cancer Res. 2008;68(19):7864–7871. doi:10.1158/0008-5472.CAN-07-6538

17. Akbar N, Azzimato V, Choudhury RP, et al. Extracellular vesicles in metabolic disease. Diabetologia. 2019;62(12):2179–2187. doi:10.1007/s00125-019-05014-5

18. Joshi BS, de Beer MA, Giepmans BNG, et al. Endocytosis of extracellular vesicles and release of their cargo from endosomes. ACS nano. 2020;14(4):4444–4455. doi:10.1021/acsnano.9b10033

19. Marostica G, Gelibter S, Gironi M, et al. Extracellular vesicles in neuroinflammation. Front Cell Dev Biol. 2020;8:623039. doi:10.3389/fcell.2020.623039

20. Jin Q, Wu P, Zhou X, et al. Extracellular vesicles: novel roles in neurological disorders. Stem Cells Int. 2021;2021:6640836. doi:10.1155/2021/6640836

21. Todorova D, Simoncini S, Lacroix R, et al. Extracellular vesicles in angiogenesis. Circ Res. 2017;120(10):1658–1673. doi:10.1161/CIRCRESAHA.117.309681

22. Willms E, Johansson HJ, Mäger I, et al. Cells release subpopulations of exosomes with distinct molecular and biological properties. Sci Rep. 2016;6:22519. doi:10.1038/srep22519

23. Kalra H, Drummen GPC, Mathivanan S. Focus on extracellular vesicles: introducing the next small big thing. Int J Mol Sci. 2016;17(2):170. doi:10.3390/ijms17020170

24. Xu X, Lai Y, Hua Z-C. Apoptosis and apoptotic body: disease message and therapeutic target potentials. Biosci Rep. 2019;39(1). doi:10.1042/BSR20180992

25. Meldolesi J. Exosomes and ectosomes in intercellular communication. Current Biol. 2018;28(8):R435–R444. doi:10.1016/j.cub.2018.01.059

26. Sharma P, Mesci P, Carromeu C, et al. Exosomes regulate neurogenesis and circuit assembly. Proc Natl Acad Sci USA. 2019;116(32):16086–16094. doi:10.1073/pnas.1902513116

27. Men Y, Yelick J, Jin S, et al. Exosome reporter mice reveal the involvement of exosomes in mediating neuron to astroglia communication in the CNS. Nat Commun. 2019;10(1):4136. doi:10.1038/s41467-019-11534-w

28. Gabrielli M, Battista N, Riganti L, et al. Active endocannabinoids are secreted on extracellular membrane vesicles. EMBO Rep. 2015;16(2):213–220. doi:10.15252/embr.201439668

29. Casella G, Rasouli J, Boehm A, et al. Oligodendrocyte-derived extracellular vesicles as antigen-specific therapy for autoimmune neuroinflammation in mice. Sci Transl Med. 2020;12(568). doi:10.1126/scitranslmed.aba0599.

30. Jin T, Gu J, Li Z, et al. Recent advances on extracellular vesicles in central nervous system diseases. Clin Interv Aging. 2021;16:257–274. doi:10.2147/CIA.S288415

31. Kalani MYS, Alsop E, Meechoovet B, et al. Extracellular microRNAs in blood differentiate between ischaemic and haemorrhagic stroke subtypes. J Extracell Vesicles. 2020;9(1):1713540. doi:10.1080/20013078.2020.1713540

32. Zheng X, Zhang L, Kuang Y, et al. Extracellular vesicles derived from neural progenitor cells–a preclinical evaluation for stroke treatment in mice. Transl Stroke Res. 2021;12(1):185–203. doi:10.1007/s12975-020-00814-z

33. Webb RL, Kaiser EE, Jurgielewicz BJ, et al. Human neural stem cell extracellular vesicles improve recovery in a porcine model of ischemic stroke. Stroke. 2018;49(5):1248–1256. doi:10.1161/STROKEAHA.117.020353

34. Xin H, Katakowski M, Wang F, et al. MicroRNA cluster miR-17-92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke. 2017;48(3):747–753.

35. Lombardi M, Parolisi R, Scaroni F, et al. Detrimental and protective action of microglial extracellular vesicles on myelin lesions: astrocyte involvement in remyelination failure. Acta Neuropathol. 2019;138(6):987–1012. doi:10.1007/s00401-019-02049-1

36. Yin Q, Ji X, Lv R, et al. Targetting exosomes as a new biomarker and therapeutic approach for Alzheimer’s disease. Clin Interv Aging. 2020;15:195–205. doi:10.2147/CIA.S240400

37. Wang P, Shao B-Z, Deng Z, et al. Autophagy in ischemic stroke. Prog Neurobiol. 2018;163–164:98–117. doi:10.1016/j.pneurobio.2018.01.001

38. Sun Y, Zhu Y, Zhong X, et al. Crosstalk between autophagy and cerebral ischemia. Front Neurosci. 2018;12:1022. doi:10.3389/fnins.2018.01022

39. Mo Y, Sun Y, Liu K. Autophagy and inflammation in ischemic stroke. Neural Regen Res. 2020;15(8):1388–1396. doi:10.4103/1673-5374.274331

40. Liu J, Guo Z-N, Yan X-L, et al. Crosstalk between autophagy and ferroptosis and its putative role in ischemic stroke. Front Cell Neurosci. 2020;14:577403. doi:10.3389/fncel.2020.577403

41. Xu W, Ocak U, Gao L, et al. Selective autophagy as a therapeutic target for neurological diseases. Cell Mol Life Sci. 2021;78(4):1369–1392. doi:10.1007/s00018-020-03667-9

42. Mizushima N, Ohsumi Y, Yoshimori T. Autophagosome formation in mammalian cells. Cell Struct Funct. 2002;27(6):421–429. doi:10.1247/csf.27.421

43. Ichimiya T, Yamakawa T, Hirano T, et al. Autophagy and autophagy-related diseases: a review. Int J Mol Sci. 2020;21(23):8974. doi:10.3390/ijms21238974

44. Li W, Li J, Bao J. Microautophagy: lesser-known self-eating. Cell Mol Life Sci. 2012;69(7):1125–1136. doi:10.1007/s00018-011-0865-5

45. Tasset I, Cuervo A. Role of chaperone-mediated autophagy in metabolism. FEBS J. 2016;283(13):2403–2413. doi:10.1111/febs.13677

46. Peker N, Gozuacik D. Autophagy as a cellular stress response mechanism in the nervous system. J Mol Biol. 2020;432(8):2560–2588. doi:10.1016/j.jmb.2020.01.017

47. Wei K, Wang P, Miao C. A double-edged sword with therapeutic potential: an updated role of autophagy in ischemic cerebral injury. CNS Neurosci Ther. 2012;18(11):879–886. doi:10.1111/cns.12005

48. Ajoolabady A, Wang S, Kroemer G, et al. Targeting autophagy in ischemic stroke: from molecular mechanisms to clinical therapeutics. Pharmacol Ther. 2021;225:107848.

49. Wang M, Lee H, Elkin K, et al. Detrimental and beneficial effect of autophagy and a potential therapeutic target after ischemic stroke. Evid Based Complement Alternat Med. 2020;2020:8372647. doi:10.1155/2020/8372647

50. Li Q, Zhang T, Wang J, et al. Rapamycin attenuates mitochondrial dysfunction via activation of mitophagy in experimental ischemic stroke. Biochem Biophys Res Commun. 2014;444(2):182–188. doi:10.1016/j.bbrc.2014.01.032

51. Nabavi S, Sureda A, Sanches-Silva A, et al. Novel therapeutic strategies for stroke: the role of autophagy. Crit Rev Clin Lab Sci. 2019;56(3):182–199. doi:10.1080/10408363.2019.1575333

52. Bar-Yosef T, Damri O, Agam G. Dual role of autophagy in diseases of the central nervous system. Front Cell Neurosci. 2019;13:196. doi:10.3389/fncel.2019.00196

53. Rubinsztein D, DiFiglia M, Heintz N, et al. Autophagy and its possible roles in nervous system diseases, damage and repair. Autophagy. 2005;1(1):11–22. doi:10.4161/auto.1.1.1513

54. Rubinsztein D, Gestwicki JE, Murphy LO, et al. Potential therapeutic applications of autophagy. Nat Rev Drug Dis. 2007;6(4):304–312. doi:10.1038/nrd2272

55. Menzies FM, Fleming A, Caricasole A, et al. Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron. 2017;93(5):1015–1034. doi:10.1016/j.neuron.2017.01.022

56. Chong Z, Shang YC, Wang S, et al. A critical kinase cascade in neurological disorders: PI 3-K, Akt, and mTOR. Future Neurol. 2012;7(6):733–748. doi:10.2217/fnl.12.72

57. Jung C, Ro S-H, Cao J, et al. mTOR regulation of autophagy. FEBS Lett. 2010;584(7):1287–1295. doi:10.1016/j.febslet.2010.01.017

58. Li W, Yu SP, Chen D, et al. The regulatory role of NF-κB in autophagy-like cell death after focal cerebral ischemia in mice. Neuroscience. 2013;244:16–30. doi:10.1016/j.neuroscience.2013.03.045

59. Cho B, Choi SY, Park O-H, et al. Differential expression of BNIP family members of BH3-only proteins during the development and after axotomy in the rat. Mol Cells. 2012;33(6):605–610. doi:10.1007/s10059-012-0051-0

60. Xin X, Pan J, Wang X-Q, et al. 2-methoxyestradiol attenuates autophagy activation after global ischemia. Can J Neurol Sci. 2011;38(4):631–638. doi:10.1017/S031716710001218X

61. Jiang T, Harder B, Rojo de la Vega M, et al. p62 links autophagy and Nrf2 signaling. Free Radic Biol Med. 2015;88:199–204. doi:10.1016/j.freeradbiomed.2015.06.014

62. Gerónimo-Olvera C, Massieu L. Autophagy as a homeostatic mechanism in response to stress conditions in the central nervous system. Mol Neurobiol. 2019;56(9):6594–6608. doi:10.1007/s12035-019-1546-x

63. Ogata M, S Hino, A Saito, et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol. 2006;26(24):9220–9231. doi:10.1128/MCB.01453-06

64. Xu J, Camfield R, Gorski SM. The interplay between exosomes and autophagy–partners in crime. J Cell Sci. 2018;131(15):jcs215210. doi:10.1242/jcs.215210

65. Auger C, Christou N, Brunel A, et al. Autophagy and extracellular vesicles in colorectal cancer: interactions and common actors? Cancers. 2021;13(5):1039. doi:10.3390/cancers13051039

66. You L, Mao L, Wei J,et al. The crosstalk between autophagic and endo-/exosomal pathways in antigen processing for MHC presentation in anticancer T cell immune responses. J Hematol Oncol. 2017;10(1):1–9.

67. Fader C, Colombo MIJA. Multivesicular bodies and autophagy in erythrocyte maturation. Autophagy. 2006;2(2):122–125. doi:10.4161/auto.2.2.2350

68. Fader CM, Sánchez D, Furlán M, et al. Induction of autophagy promotes fusion of multivesicular bodies with autophagic vacuoles in k562 cells. Traffic. 2008;9(2):230–250.

69. Alvarez-Erviti L, Seow Y, Schapira AH, et al. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol Dis. 2011;42(3):360–367. doi:10.1016/j.nbd.2011.01.029

70. Fussi N, Höllerhage M, Chakroun T, et al. Exosomal secretion of alpha-synuclein as protective mechanism after upstream blockage of macroautophagy. Cell Death Dis. 2018;9(7):757. doi:10.1038/s41419-018-0816-2

71. Liu J, Liu C, Zhang J, et al. A self-assembled alpha-synuclein nanoscavenger for parkinson’s disease. ACS Nano. 2020;14(2):1533–1549. doi:10.1021/acsnano.9b06453

72. Willen K, Edgar JR, Hasegawa T, et al. Abeta accumulation causes MVB enlargement and is modelled by dominant negative VPS4A. Mol Neurodegener. 2017;12(1):61. doi:10.1186/s13024-017-0203-y

73. Xin H, Katakowski M, Wang F, et al. Correction to: microRNA-17-92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke. 2017;48(5):e137. doi:10.1161/STR.0000000000000130

74. Ling D, Magallanes M, Salvaterra PM. Accumulation of amyloid-like Aβ 1–42 in AEL (Autophagy–endosomal–lysosomal) vesicles: potential implications for plaque biogenesis. ASN Neuro. 2014;6(2):AN20130044. doi:10.1042/AN20130044

75. Saman S, Lee NCY, Inoyo I, et al. Proteins recruited to exosomes by tau overexpression implicate novel cellular mechanisms linking tau secretion with Alzheimer’s disease. J Alzheimers Dis. 2014;40 Suppl 1:S47–S70. doi:10.3233/JAD-132135

76. Ahmed ME, Iyer S, Thangavel R, et al. Co-localization of Glia maturation factor with NLRP3 inflammasome and autophagosome markers in human Alzheimer’s disease brain. J Alzheimers Dis. 2017;60(3):1143–1160.

77. Houtman J, Freitag K, Gimber N, et al. Beclin1-driven autophagy modulates the inflammatory response of microglia via NLRP 3. EMBO J. 2019;38(4):e99430.

78. Kong SM, Chan BK, Park JS, et al. Parkinson’s disease-linked human PARK9/ATP13A2 maintains zinc homeostasis and promotes alpha-Synuclein externalization via exosomes. Hum Mol Genet. 2014;23(11):2816–2833. doi:10.1093/hmg/ddu099

79. Danzer KM, Kranich LR, Ruf WP, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegener. 2012;7(1):1–18.

80. Guo M, Wang J, Zhao Y, et al. Microglial exosomes facilitate α-synuclein transmission in Parkinson’s disease. Brain. 2020;143(5):1476–1497.

81. Doeppner TR, Herz J, Görgens A, et al. Extracellular vesicles improve post‐stroke neuroregeneration and prevent postischemic immunosuppression. Stem Cells Transl Med. 2015;4(10):1131–1143. doi:10.5966/sctm.2015-0078

82. Song Y, Li Z, He T, et al. M2 microglia-derived exosomes protect the mouse brain from ischemia-reperfusion injury via exosomal miR-124. Theranostics. 2019;9(10):2910. doi:10.7150/thno.30879

83. Yang J, Zhang X, Chen X, et al. Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia. Mol Ther Nucleic Acids. 2017;7:278–287. doi:10.1016/j.omtn.2017.04.010

84. Shen H, Yao X, Li H, et al. Role of exosomes derived from miR-133b modified MSCs in an experimental rat model of intracerebral hemorrhage. J Mol Neurosci. 2018;64(3):421–430. doi:10.1007/s12031-018-1041-2

85. Xia Y, Ling X, Hu G, et al. Small extracellular vesicles secreted by human iPSC-derived MSC enhance angiogenesis through inhibiting STAT3-dependent autophagy in ischemic stroke. Stem cell Res Ther. 2020;11(1):1–17.

86. Wang W, Li D-B, Li R-Y, et al. Diagnosis of hyperacute and acute ischaemic stroke: the potential utility of exosomal MicroRNA-21-5p and MicroRNA-30a-5p. Cerebrovasc Dis. 2018;45(5–6):204–212. doi:10.1159/000488365

87. Yang H-C, Zhang M, Wu R, et al. CC chemokine receptor type 2-overexpressing exosomes alleviated experimental post-stroke cognitive impairment by enhancing microglia/macrophage M2 polarization. World J Stem Cells. 2020;12(2):152.

88. Pei X, Li Y, Zhu L, et al. Astrocyte-derived exosomes suppress autophagy and ameliorate neuronal damage in experimental ischemic stroke. Exp Cell Res. 2019;382(2):111474. doi:10.1016/j.yexcr.2019.06.019

89. Zang J, Wu Y, Su X, et al. Inhibition of PDE1-B by vinpocetine regulates microglial exosomes and polarization through enhancing autophagic flux for neuroprotection against ischemic stroke. Front Cell Dev Biol. 2020;8:616590.

90. Chen W, Wang H, Zhu Z, et al. Exosome-Shuttled circSHOC2 from IPASs regulates neuronal autophagy and ameliorates ischemic brain injury via the miR-7670-3p/SIRT1 axis. Mol Ther Nucleic Acids. 2020;22:657–672.

91. Pei X, Li Y, Zhu L, et al. Astrocyte-derived exosomes transfer miR-190b to inhibit oxygen and glucose deprivation-induced autophagy and neuronal apoptosis. Cell Cycle. 2020;19(8):906–917.

92. Bu X, Li D, Wang F, et al. Protective role of astrocyte-derived exosomal microRNA-361 in cerebral ischemic-reperfusion injury by regulating the AMPK/mTOR signaling pathway and targeting CTSB. Neuropsychiatr Dis Treat. 2020;16:1863–1877. doi:10.2147/NDT.S260748

93. Jiang Y, Xie H, Tu W, et al. Exosomes secreted by HUVECs attenuate hypoxia/reoxygenation-induced apoptosis in neural cells by suppressing miR-21-3p. Am J Transl Res. 2018;10(11):3529.

94. Gallart-Palau X, Guo X, Serra A, et al. Alzheimer’s disease progression characterized by alterations in the molecular profiles and biogenesis of brain extracellular vesicles. Alzheimers Res Ther. 2020;12(1):54. doi:10.1186/s13195-020-00623-4

95. Scesa G, Moyano AL, Bongarzone ER, et al. Port-to-port delivery: mobilization of toxic sphingolipids via extracellular vesicles. J Neurosci Res. 2016;94(11):1333–1340. doi:10.1002/jnr.23798

96. Xu Z, Nan W, Zhang X, et al. Umbilical cord mesenchymal stem cells conditioned medium promotes Abeta25-35 phagocytosis by modulating autophagy and abeta-degrading enzymes in BV2 cells. J Mol Neurosci. 2018;65(2):222–233. doi:10.1007/s12031-018-1075-5

97. Cui G-H, Wu J, Mou F-F, et al. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J. 2018;32(2):654–668. doi:10.1096/fj.201700600R

98. Xia Y, Zhang G, Han C, et al. Microglia as modulators of exosomal alpha-synuclein transmission. Cell Death Dis. 2019;10(3):174. doi:10.1038/s41419-019-1404-9

99. Zhou T, Lin D, Chen Y, et al. α-synuclein accumulation in SH-SY5Y cell impairs autophagy in microglia by exosomes overloading miR-19a-3p. Epigenomics. 2019;11(15):1661–1677.

100. Li Q, Wang Z, Xing H, et al. Exosomes derived from miR-188-3p-modified adipose-derived mesenchymal stem cells protect Parkinson’s disease. Mol Ther Nucleic Acids. 2021;23:1334–1344.

101. Fan RZ, Guo M, Luo S, et al. Exosome release and neuropathology induced by alpha-synuclein: new insights into protective mechanisms of Drp1 inhibition. Acta Neuropathol Commun. 2019;7(1):184. doi:10.1186/s40478-019-0821-4

102. Kim MJ, Deng H-X, Wong YC, et al. The Parkinson’s disease-linked protein TMEM230 is required for Rab8a-mediated secretory vesicle trafficking and retromer trafficking. Hum Mol Genet. 2017;26(4):729–741. doi:10.1093/hmg/ddw413

103. Underwood R, Wang B, Carico C, et al. The GTPase Rab27b regulates the release, autophagic clearance, and toxicity of alpha-synuclein. J Biol Chem. 2020;295(23):8005–8016. doi:10.1074/jbc.RA120.013337

104. Harischandra DS, Ghaisas S, Rokad D, et al. Environmental neurotoxicant manganese regulates exosome-mediated extracellular miRNAs in cell culture model of Parkinson’s disease: relevance to alpha-synuclein misfolding in metal neurotoxicity. Neurotoxicology. 2018;64:267–277. doi:10.1016/j.neuro.2017.04.007

105. Li D, Huang S, Zhu J, et al. Exosomes from MiR-21-5p-increased neurons play a role in neuroprotection by suppressing Rab11a-mediated neuronal autophagy in vitro after traumatic brain injury. Med Sci Monitor. 2019;25:1871.

106. Li D, Huang S, Yin Z, et al. Increases in miR-124-3p in microglial exosomes confer neuroprotective effects by targeting FIP200-mediated neuronal autophagy following traumatic brain injury. Neurochem Res. 2019;44(8):1903–1923.

107. Wang P, Ma H, Zhang Y, et al. Plasma exosome-derived microRNAs as novel biomarkers of traumatic brain injury in rats. Int J Med Sci. 2020;17(4):437. doi:10.7150/ijms.39667

108. Liu Q, Deng J, Qiu Y, et al. Non-coding RNA basis of muscle atrophy. Mol Ther Nucleic Acids. 2021;26:1066–1078. doi:10.1016/j.omtn.2021.10.010

109. Liao Y, Guo S, Liu G, et al. Host non-coding RNA regulates influenza A virus replication. Viruses. 2021;14(1):51. doi:10.3390/v14010051

110. Xin W, Wei W, Pan Y-L, et al. Modulating poststroke inflammatory mechanisms: novel aspects of mesenchymal stem cells, extracellular vesicles and microglia. World J Stem Cells. 2021;13(8):1030–1048. doi:10.4252/wjsc.v13.i8.1030

111. Ge X, Lei P, Wang H-C, et al. miR-21 improves the neurological outcome after traumatic brain injury in rats. Sci Rep. 2014;4:6718. doi:10.1038/srep06718

112. Ge X, Han Z, Chen F, et al. MiR-21 alleviates secondary blood-brain barrier damage after traumatic brain injury in rats. Brain Res. 2015;1603:150–157. doi:10.1016/j.brainres.2015.01.009

113. Liu Y, Li Y-P, Xiao L-M, et al. Extracellular vesicles derived from M2 microglia reduce ischemic brain injury through microRNA-135a-5p/TXNIP/NLRP3 axis. Lab Invest. 2021;101(7):837–850. doi:10.1038/s41374-021-00545-1

114. Gulati S, Palczewski K, Engel A, et al. Cryo-EM structure of phosphodiesterase 6 reveals insights into the allosteric regulation of type I phosphodiesterases. Sci Adv. 2019;5(2):eaav4322. doi:10.1126/sciadv.aav4322

115. Akira S, Nishio Y, Inoue M, et al. Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway. Cell. 1994;77(1):63–71. doi:10.1016/0092-8674(94)90235-6

116. You L, Wang Z, Li H, et al. The role of STAT3 in autophagy. Autophagy. 2015;11(5):729–739. doi:10.1080/15548627.2015.1017192

117. Shen S, Niso-Santano M, Adjemian S, et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity. Mol Cell. 2012;48(5):667–680. doi:10.1016/j.molcel.2012.09.013

118. Du Toit A. Autophagy: STAT3 maintains order. Nat Rev Mol Cell Biol. 2012;13(12):754. doi:10.1038/nrm3472

119. Yokoyama T, Kondo Y, Kondo S. Roles of mTOR and STAT3 in autophagy induced by telomere 3’ overhang-specific DNA oligonucleotides. Autophagy. 2007;3(5):496–498. doi:10.4161/auto.4602

120. Vassileff N, Cheng L, Hill AF. Hill, Extracellular vesicles–propagators of neuropathology and sources of potential biomarkers and therapeutics for neurodegenerative diseases. J Cell Sci. 2020;133(23):jcs243139. doi:10.1242/jcs.243139

121. Zheng J, Tan J, Miao -Y-Y, et al. Extracellular vesicles degradation pathway based autophagy lysosome pathway. Am J Transl Res. 2019;11(3):1170.

Creative Commons License © 2022 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.