Skip to main content

MINI REVIEW article

Front. Cell. Neurosci., 19 March 2024
Sec. Cellular Neuropathology
This article is part of the Research Topic Extracellular vesicles: emerging roles in the aged and neurodegenerative brain View all articles

Engineered exosomes enriched with select microRNAs amplify their therapeutic efficacy for traumatic brain injury and stroke

  • 1Department of Neurosurgery, Henry Ford Health, Detroit, MI, United States
  • 2Department of Neurology, Henry Ford Health, Detroit, MI, United States
  • 3Department of Physics, Oakland University, Rochester, MI, United States

Traumatic brain injury (TBI) and stroke stand as prominent causes of global disability and mortality. Treatment strategies for stroke and TBI are shifting from targeting neuroprotection toward cell-based neurorestorative strategy, aiming to augment endogenous brain remodeling, which holds considerable promise for the treatment of TBI and stroke. Compelling evidence underscores that the therapeutic effects of cell-based therapy are mediated by the active generation and release of exosomes from administered cells. Exosomes, endosomal derived and nano-sized extracellular vesicles, play a pivotal role in intercellular communication. Thus, we may independently employ exosomes to treat stroke and TBI. Systemic administration of mesenchymal stem cell (MSC) derived exosomes promotes neuroplasticity and neurological functional recovery in preclinical animal models of TBI and stroke. In this mini review, we describe the properties of exosomes and recent exosome-based therapies of TBI and stroke. It is noteworthy that the microRNA cargo within exosomes contributes to their therapeutic effects. Thus, we provide a brief introduction to microRNAs and insight into their key roles in mediating therapeutic effects. With the increasing knowledge of exosomes, researchers have “engineered” exosome microRNA content to amplify their therapeutic benefits. We therefore focus our discussion on the therapeutic benefits of recently employed microRNA-enriched engineered exosomes. We also discuss the current opportunities and challenges in translating exosome-based therapy to clinical applications.

1 Introduction

Traumatic brain injury (TBI) and stroke are prominent causes of global disability and mortality, and impose a significant social and economic burden (Hyder et al., 2007; Lackland et al., 2014; Johnson and Griswold, 2017; Kuriakose and Xiao, 2020; Hering and Shetty, 2023). Unfortunately, no effective drugs are available for improving TBI functional recovery, and tPA is the only FDA-approved drug as a treatment for acute ischemic stroke (National Institute of Neurological Disorders and Stroke rt, PA Stroke Study Group, 1995; Hacke et al., 2008). Furthermore, nearly all the phase II/III clinical trials directed towards neuroprotection for TBI and stroke have failed (Xiong et al., 2018).

Recently, attention has shifted towards cell-based neurorestorative strategy, designed to augment endogenous brain remodeling after TBI or stroke. Cell-based therapy, particularly using bone marrow mesenchymal cells (MSCs), has proven to be safe and effective in promoting neuroplasticity and neurorestoration, leading to the improvement of neurological and cognitive function in animal models of TBI and stroke (Chen et al., 2001; Chopp and Li, 2002; Zhang and Chopp, 2009; Cox et al., 2011; Nichols et al., 2013; Bonsack et al., 2020; Pischiutta et al., 2022; Giovannelli et al., 2023). Paracrine effects rather than the direct replacement of injured tissue through stem/progenitor cell differentiation underlie the therapeutic benefits of cell-based therapy (Chopp and Li, 2002; Phinney and Prockop, 2007; Lai et al., 2011; Camussi et al., 2013; Xin et al., 2013a; Pischiutta et al., 2022; Giovannelli et al., 2023; Zhuang et al., 2023). Signaling pathways in the injured brain triggered by paracrine factors released directly or indirectly by MSCs have the potential to promote endogenous neuronal rewiring and enhance angiogenesis and neurogenesis by communication with brain parenchymal cells, and thereby amplify brain remodeling, and ultimately improve functional recovery after injury (Zhang et al., 2019; Pischiutta et al., 2022; Zhuang et al., 2023).

Among these paracrine factors, exosomes, endosome-derived membrane-bound small extracellular vesicles, play a significant role in the intercellular communication (Lai and Breakefield, 2012; Rak, 2013; Lener et al., 2015; Xin et al., 2021; Zhang Y. et al., 2021, 2023; Liu et al., 2022). Exosomes contain proteins, lipids, mRNAs, microRNAs (miRNAs), and long non-coding RNAs. Through the transfer of their molecules via endocytosis or ligand-receptor interactions, exosomes communicate directly or indirectly with endogenous brain cells (Schneider and Simons, 2013). This communication may promote neurorestorative effects and improve functional outcome after TBI or stroke (Xin et al., 2013a, 2014; Kim et al., 2016; Zhang et al., 2017b). Contained within the exosome cargo, miRNAs are of considerable importance in mediating the therapeutic effects of exosomes (Xin et al., 2013a, 2014; Zhang et al., 2019).

In this mini review, we discuss exosome-based treatment as a potential neurorestorative therapy for TBI and stroke. We focus on recent developments of engineered exosome-based treatment, especially specific miRNA enriched exosomes. At the end of this mini review, we discuss the current opportunities and challenges in translation of exosome-based therapy to clinical applications.

2 Exosomes properties and physiological functions

Exosomes are membrane-bound vesicles derived from endosomes, typically measuring ~30–150 nm in diameter (Lai and Breakefield, 2012; Rak, 2013). The formation of exosomes is initiated through cellular endocytosis or plasma membrane invagination, leading to the generation of small intracellular bodies called endosomes (Thery et al., 2002). Early endosomes subsequently develop into late endosomes, which contain numerous intraluminal vesicles (ILVs) and is often referred to as a multivesicular body (MVB). Proteins, mRNAs, miRNAs, and DNAs are directly sorted to the MVB from several organelles (Thery et al., 1999). Later, the MVB may either fuse with the lysosome, resulting in the degradation of its contents, or fuse with the plasma membrane, leading to the release of its ILVs into extracellular environment. These vesicles are then called exosomes (van Niel et al., 2006).

The cargos and membrane structure of exosomes are determined by their birth cells under specific physiological and environmental conditions of these cells (Chopp and Zhang, 2015). Under physiological conditions, via transfer of their cargo, exosomes, secreted by brain cells, maintain or regulate brain function (Kalluri and LeBleu, 2020). For example, exosomes derived from neurons maintain the integrity of the blood–brain barrier (BBB) by transferring miRNA-132 to endothelial cells (Xu et al., 2017); oligodendrocyte derived exosomes assist in axonal myelination by delivering myelin proteins proteolipid protein (PLP), 2′3’-cyclic-nucleotide-phosphodiesterase (CNP) and myelin basic protein (MBP) (Kramer-Albers et al., 2007; Fruhbeis et al., 2013a,b); and astrocyte-derived exosomes regulate synaptic plasticity by transporting the miRNA-26 to synapses (Lafourcade et al., 2016).

The mechanisms underlying exosome treatment for brain injury involve complex intercellular communication and the delivery of bioactive molecules to target cells within the injured brain tissue. Exosomes possess anti-inflammatory properties that suppress neuroinflammation in the injured brain (Zhang et al., 2015; Yang Y. et al., 2017; Williams et al., 2020a; Mavroudis et al., 2023). Interacting with immune cells in the brain, such as microglia and astrocytes, exosomes modulate the immune response following injury (Liu et al., 2023). Exosomes exert neuroprotective effects by promoting cell survival and inhibiting apoptosis of injured neurons (Williams et al., 2020a; Zhang et al., 2022). Treatment with exosomes derived from stem cells stimulate angiogenesis (formation of new blood vessels) and neurogenesis (generation of new neurons) in the injured brain (Zhang et al., 2015). In addition, exosomes influence the integrity and permeability of the BBB and play a role in promoting neuronal plasticity by delivering factors that modulate synaptic function and neuronal connectivity (Gao et al., 2018; Williams et al., 2020a; Xia et al., 2022). Overall, exosome therapy for brain injury harnesses a range of molecular mechanisms and pathways to promote neuroprotection, tissue repair, and functional recovery. By capitalizing on the therapeutic potential of exosomes, researchers aim to develop effective treatments for TBI, stroke, and neurodegenerative diseases. A detailed overview of the therapeutic effects and mechanisms underlying naive and engineered exosome treatment for TBI and stroke is provided in following Sections, with a focus on the role of miRNAs in mediating exosome function.

3 Exosome-based therapy of stroke and TBI

Paracrine mechanisms underlie the MSC-based therapeutic effects, where MSCs secrete factors that influence endogenous cells (Chopp and Li, 2002; Lai et al., 2011; Camussi et al., 2013; Xin et al., 2013a). Among these paracrine factors, exosomes are critical to cell-based therapeutic actions (Xin et al., 2014; Yang Y. et al., 2017; Xiong et al., 2018; Zhang et al., 2019). Compared to cell-based therapy, exosome-based therapy offers several advantages: (1) exosomes have a superior safety profile due to low immunogenicity and tumorigenesis (El Andaloussi et al., 2013; Xiong et al., 2017); (2) exosome injection has low risk of inducing microvascular embolism due to its nano size (Xin et al., 2014); (3) exosomes can be safely stored without losing function; (4) nano-sized exosomes are capable of crossing the BBB by systemic injection (Chen et al., 2016; Otero-Ortega et al., 2018); (5) the cargo of exosomes can be engineered to amplify their therapeutic effects (Xin et al., 2017b; Chen and Chopp, 2018).

Pioneering research on MSC-exosome-based treatment has been performed in rodent models of stroke and TBI (Xin et al., 2013a; Doeppner et al., 2015; Zhang et al., 2015). In the stroke study, systemic administration of MSC-exosomes (MSC-Exos) in rats 24 h after induction of middle cerebral artery occlusion (MCAO) improved their neurological functional recovery, possibly by enhancing neurite and vascular remodeling and neurogenesis post stroke (Xin et al., 2013a). In the TBI study, MSC-Exos administered intravenously 24 h after TBI promoted neurovascular remodeling, neurogenesis, and sensorimotor and cognitive functional recovery in TBI rats (Zhang et al., 2015). Importantly, MSC-Exos and MSC treatments resulted in equivalent functional improvements in mice post-stroke (Doeppner et al., 2015). Increasing numbers preclinical studies have shown that exosome-based treatment can promote neuroplasticity and neurorestoration by increasing neurite remodeling (Xin et al., 2013a; Otero-Ortega et al., 2017), axonal sprouting (Otero-Ortega et al., 2017; Xin et al., 2017a), synaptogenesis (Xin et al., 2017a), and angiogenesis and neurogenesis (Zhang et al., 2020), ultimately improving functional outcomes in stroke and TBI animals (Zhang et al., 2017b, 2020; Dumbrava et al., 2022; Zhang R. et al., 2023). MSC-Exos also promoted neurological functional recovery and brain tissue remodeling in aged stroke rats (Dumbrava et al., 2022). In addition to neurorestorative effects, exosomes have neuroprotective effects, such as reducing lesion volume in TBI (Zhang W. et al., 2021) or ischemic core (Liu et al., 2021), suppressing cell apoptosis (Zhang W. et al., 2021), and mitigating neuroinflammation (Zhang R. et al., 2023). These preclinical data demonstrated the great therapeutic potential of exosome-based therapy for stroke and TBI.

Following the rodent studies, exosome-based therapy has been further evaluated in large animal models. In a swine model of severe TBI and hemorrhagic shock (HS), early (1 h after shock)-single-dose MSC-Exos-treated animals experienced significantly less functional impairment and faster neurological functional recovery, resulting from reduced brain lesion, inflammation, and apoptosis, as well as promoted neural plasticity (Williams et al., 2020b). In a series of studies using an adult rhesus monkey model with cortical injury, MSC-Exos treatment achieved increased functional recovery of grasp pattern with reduced latency to retrieve a food reward compared to saline treatment (Moore et al., 2019; Go et al., 2020, 2021). These therapeutic effects were attributed to the neuroprotection and neurorestoration effects of MSC-Exos, by reducing neuroinflammation (Go et al., 2020) and suppressing damage to oligodendrocytes to improve the myelin maintenance (Go et al., 2021). This cortical injury model in the monkey is highly related to stroke in human due to the similarity of fine motor function of hand and digits between human and monkey.

In addition to MSCs, exosomes derived from other cell types also induce neurorestoration and improve functional outcomes. For example, neural stem cell (NSC)-derived exosomes reduced inflammatory response, ameliorated brain injury, and improved motor functional recovery in stroke mice (Zhang R. et al., 2023); astrocyte-derived exosomes protected mice and rats against TBI-induced neuronal cell loss/apoptosis and oxidative stress, thereby alleviating functional impairment (Zhang W. et al., 2021). The cargos of exosomes are closely related to their parent cells and mediate their therapeutic effects in stroke and TBI treatment (Zhang et al., 2019). However, the most effective types of cell-derived exosomes for the treatment of stroke or TBI have not yet been determined (Lener et al., 2015).

In summary, the data from rodent and large animal studies suggest that exosome-based therapy provides beneficial therapeutic effects post stroke and TBI, including neurorestoration and neuroprotection (Table 1).

Table 1
www.frontiersin.org

Table 1. Selected recent studies of exosomes for treatment of stroke and TBI.

4 MiRNAs in exosomes are critical to therapeutic effects

As noted, exosomes can alter recipient cells’ function via transfer of their cargo. Systemic administration of liposomes, consisting of lipid components of exosomes but without proteins and genetic materials, had no therapeutic benefits compared to naïve MSC-Exos treatment, indicating that the therapeutic effects of exosomes are derived from the cargo of exosomes (Zhang et al., 2017b). Among the exosomal cargo, miRNAs play a substantial role in therapeutic effects of exosomes.

MiRNAs are small (22 nucleotides in length), evolutionary conserved, non-coding RNA molecules (Macfarlane and Murphy, 2010; O'Brien et al., 2018). The biogenesis of most miRNAs is initiated from the transcription of a DNA sequence, and transcribed primary miRNAs are subsequently processed into precursor miRNAs and mature miRNAs (O'Brien et al., 2018). Each miRNA is a master molecular switch, which regulates the gene expression of hundreds of mRNAs at post-transcriptional level by binding to complementary sequences on mRNA, consequently causes the cleavage of mRNAs or translation repression (Cai et al., 2009; Macfarlane and Murphy, 2010).

Through genetic approaches, many studies have demonstrated that miRNAs contribute to the therapeutic effect of exosomes (Mateescu et al., 2017). Dicer, a ribonuclease, is involved in the production of mature miRNAs. Conditional knockout of Dicer (Dicer/Cko) in adult neural progenitor cells (NPCs) substantially reduced cellular miRNAs, and impaired neurogenesis and cognitive function in Dicer/Cko mice, while administration of cerebral endothelial-derived exosomes carrying mature miRNAs restored neurogenesis and cognitive function (Zhang, R. L. et al., 2017). Another example is Argonaute 2 (Ago2), a primary miRNA machinery protein required for packaging miRNAs into exosomes and performing activities in the recipient cells (Gibbings et al., 2009). MSC-Exos promoted the axonal growth for cortical neurons while attenuation of Ago2 protein in MSC-Exos abolished their effect on axonal growth (Zhang et al., 2017a). Attenuation of Ago2 protein in MSCs reduces miRNAs in MSC-Exos and reduces exosome treatment-induced beneficial effects in TBI recovery (Zhang Y. et al., 2023). Collectively, these studies indicate that therapeutic benefits of exosomes are substantially attributed to their miRNA cargo.

5 Engineered exosomes to amplify therapeutic benefits

Since miRNAs significantly contribute to the exosome’s therapeutic benefits, the use of engineered exosomes with enriched specific miRNAs for the treatment of stroke and TBI is under active investigation in order to amplify their therapeutic benefits. Generally, designing miRNA enriched engineered exosomes begins with brain miRNA profiling after stroke or TBI (Deng et al., 2019; Zhuang et al., 2023). Dysregulation of miRNA expression is related to neurodegenerative diseases and brain injuries, including stroke and TBI (Pan et al., 2017; Atif and Hicks, 2019). MiRNA microarray and next-generation sequencing platform are employed to quantify miRNA expression profiles in brain tissue, blood, and cerebrospinal fluid of animal models and humans with stroke or TBI (Redell et al., 2010; Di Pietro et al., 2017; Pan et al., 2017). After comparing these miRNA profiles with those from healthy animals or humans, researchers apply bioinformatic analysis to identify specific microRNAs that play crucial roles in neuroprotection, neuroregeneration, and anti-inflammatory responses, and anticipate the candidate gene targets of these miRNAs to determine if they may be a target for stroke or TBI treatment (Zhuang et al., 2023). Then, researchers tailor the composition of engineered exosomes to carry these microRNAs for precise targeting of pathways involved in TBI and stroke recovery by altering the genetic character of cells, e.g., by transfection or electroporation miRNA mimics (agomir) / inhibitors (antagomir), finally generating specific miRNA enriched/decreased engineered exosomes (Wen, 2016; Sun et al., 2018). Recently published research about miRNAs enriched exosomes for the treatment of brain diseases is listed in Table 2.

Table 2
www.frontiersin.org

Table 2. Studies of miRNA-enriched exosomes for treatment of neural injuries.

The therapeutic potential of miRNAs enriched engineered exosomes derived from MSCs in stroke rodent model was first documented in 2013 (Xin et al., 2013b). In 2017, three engineered exosomes with elevated miRNAs were developed and provided an increased therapeutic effect on neurological recovery compared with naïve exosomes (Xin et al., 2017a,b; Yang J. et al., 2017). Systemic administration of MSC-Exo-17-92 in a rat MCAO model at 24 h after induction of stroke significantly improved sensorimotor functional recovery and enhanced neurogenesis, neurite plasticity, and oligodendrogenesis compared to naïve exosome treatment. This enhanced therapeutic benefit was attributed to downregulation of phosphatase and tension homolog (PTEN), and subsequent activation of the P13K/phosphorylated mammalian target of rapamycin (mTOR) signaling pathways targeted by the miRNA-17-92 cluster (Xin et al., 2017a). In another rodent study, MSC-Exo-133b treatment increased secondary release of exosomes from astrocytes and promoted neurite outgrowth and plasticity and functional recovery in stroke rats (Xin et al., 2017b). MSC-Exo with elevated miRNA-124 (MSC-Exo-124) with rabies virus glycoprotein (RVG) fused to the exosomal protein lysosome-associated membrane glycoprotein 2b (Lamp2b) delivered miRNA-124 more efficiently to the infarct site, and further promoted cortical neurogenesis in a mouse model of ischemic stroke (Yang J. et al., 2017).

Currently, select exosomal miRNAs from different cell lines have been verified to mediate neurorestorative function via promoting neurogenesis, angiogenesis, axonal remodeling, and neuronal plasticity (Wang et al., 2020; Nasirishargh et al., 2021). For example, miRNA-126 can regulate vascular integrity and promote angiogenesis via activating vascular endothelial growth factor receptor 2 (VEGFR2) (Wu et al., 2016). In a diabetic stroke mouse model, miRNA-126 enriched exosomes derived from endothelial progenitor cells (EPC-Exo-126) were rapidly taken up by brain neurons, endothelial cells, astrocytes, and microglia in the peri-infarct area after treatment, which more effectively promoted neurogenesis and angiogenesis than the naïve EPC-Exo, and enhanced functional recovery post-stroke (Wang et al., 2020). Engineered exosomes have also been applied to TBI treatment via boosting neurorestoration. Intravenous administration of MSC-Exo-17-92 to rat after 24 h of TBI significantly increased neurogenesis and angiogenesis in the hippocampus, which promoted neuronal functional recovery (Zhang Y. et al., 2021); MSC-Exo-124 elevated hippocampus neuronal proliferation and differentiation after TBI in rats, possibly by inhibiting the inflammation via regulating the TLR4 pathway (Yang et al., 2019).

Many exosomal miRNAs also play a neuroprotective role in stroke and TBI (Panaro et al., 2020). MiR-124, the most highly expressed miRNA in the central nervous system (CNS), is significantly altered in the acute, sub-acute and chronic phase post-TBI (Li et al., 2019; Ge et al., 2020; Zhuang et al., 2023). Microglia derived Exo-124-3p (Microglia-Exo-124-3p) alleviated neurodegeneration by reducing neurite branch loss and inhibiting β-amyloid (Aβ) by targeting the Rela/ApoE signaling pathway in a mouse repetitive (r)TBI (Ge et al., 2020); MSCs-derived Exo-124-3p (MSCs-Exo-124-3p) reduced neuronal cell death and minimized the lesion volume post-TBI in rats likely via attenuating posttraumatic glutamate-mediated excitotoxicity by downregulating p38MAPK expression (Zhuang et al., 2023). Additionally, engineered exosomes, e.g., MSC-Exo enriched with miRNA-138-5p (MSC-Exo-138-5p), miRNA-145, (MSC-Exo-145), miRNA-21-5p (MSC-Exo-21-5p), and adipose-derived stem (ADSCs) derived exosomes enriched with miRNA-30d-5p (ADSC-Exo-30d-5p) provided neuroprotection after neural injury (Jiang et al., 2018; Deng et al., 2019; Gao et al., 2020; Zhou et al., 2022). For example, the treatment of MSC-Exo-138-5p reduced apoptosis of astrocytes in ischemic stroke mice (Deng et al., 2019); ADSC-Exo-30d-5p significantly reduced infarct volume and neuronal apoptosis in stroke rats (Jiang et al., 2018).

Chronic neuroinflammation aggravates neurodegeneration and impedes neuronal repair after stroke and TBI (Xiong et al., 2018). Several miRNA-enriched exosomes are designed to regulate neuroinflammation. MSC-Exo-145 promoted the conversion of microglia from a pro-inflammatory M1 to an anti-inflammatory M2 phenotype in vitro, possibly by downregulating FOXO1 (Zhou et al., 2022). ADSC-Exo-30d-5p regulated the neuroinflammation by suppressing autophagy, M1 polarization of microglial cells, and inflammatory cytokines in vitro and in vivo, which reduced infarct size in stroke rats (Jiang et al., 2018). Moreover, MSC-Exo- and microglia-Exo-124 treatment promoted M2 polarization of microglia, and inhibited autophagy and inflammatory cytokines in a TBI rodent model (Li et al., 2019; Yang et al., 2019).

Looking forward, miRNA-enriched engineered exosomes could be further modulated to enhance treatment efficiency and amplify therapeutic effects. For example, miRNA-enriched exosomes could incorporate targeting strategies to enable site-specific drug delivery; engineered exosomes could carry multiple miRNAs to target different injury mechanisms after neural injury.

In summary, these preclinical data suggest that modulating miRNAs content of exosomes is a feasible and promising means to amplify the therapeutic effects of exosomes for the treatment of stroke and TBI, as well as degenerative diseases.

6 Discussion

Exosomes play a substantial role in intercellular communication. By transferring their cargo, exosomes can induce neurorestorative and neuroprotective effects via regulating genes and protein expression in target cells or tissues post-injury. Collectively, data from preclinical studies indicate that exosome-based therapy could promote neuroplasticity, reduce impairments, and accelerate functional recovery in animal models of stroke or TBI. Although the mechanisms that underlie the benefits are not fully understood, exosome-based approach as potential therapy for stroke and TBI is under active investigation (Zhang et al., 2019). Engineered exosomes with modified cargos are designed to amplify the therapeutic efficacy, in which specific miRNAs enriched engineered exosomes stand in the spotlight and provide better therapeutic efficacy than naïve exosomes.

Although the data from preclinical proof-concept studies are promising, there are several challenges in the translation of exosome-based therapy to clinical application. Firstly, a greater understanding of the mechanism of exosomes action is needed. This will secure the safety of exosome-based therapy and be the foundation of designed engineered exosomes. Secondly, standardization of exosome isolation, scale-up production, characterization, and cargo analysis methods are necessary for human clinical trials (Lener et al., 2015). Thirdly, the determination of optimal dose, therapeutic windows and cell sources of exosomes are crucial for successful clinical translation (Xiong et al., 2017). Performance of safety studies for using exosomes must be fully investigated, such as oncogenic potential studies (Jayaraman et al., 2017; Latchana et al., 2017; Zhang H. et al., 2017). It is because many miRNAs also are closely linked with oncogenesis (Chen et al., 2013; Zhang H. et al., 2017). Therefore, studies should be performed to ensure that the restorative exosomes would not further induce tumor growth. Collectively, further preclinical studies of cell source selection, scale-up production, safety, dose–response, time window, administration routes, cargo analyses, and mechanisms of naive and engineered exosomes are required before effective and safe clinical translation.

Author contributions

LC: Conceptualization, Writing – original draft, Writing – review & editing. YX: Conceptualization, Funding acquisition, Supervision, Writing – review & editing. MC: Conceptualization, Funding acquisition, Supervision, Writing – review & editing. YZ: Conceptualization, Funding acquisition, Supervision, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported in-part by National Institutes of Health grants, R01HL143432 (to MC) and R01NS100710 (to YX). This work was also supported by internal funding from HFH, A10271 (to YZ).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Atif, H., and Hicks, S. D. (2019). A review of MicroRNA biomarkers in traumatic brain injury. J. Exp. Neurosci. 13:117906951983228. doi: 10.1177/1179069519832286

Crossref Full Text | Google Scholar

Bonsack, B., Corey, S., Shear, A., Heyck, M., Cozene, B., Sadanandan, N., et al. (2020). Mesenchymal stem cell therapy alleviates the neuroinflammation associated with acquired brain injury. CNS Neurosci. Ther. 26, 603–615. doi: 10.1111/cns.13378

PubMed Abstract | Crossref Full Text | Google Scholar

Cai, Y., Yu, X., Hu, S., and Yu, J. (2009). A brief review on the mechanisms of miRNA regulation. Genomics Proteomics Bioinformatics 7, 147–154. doi: 10.1016/S1672-0229(08)60044-3

PubMed Abstract | Crossref Full Text | Google Scholar

Camussi, G., Deregibus, M. C., and Cantaluppi, V. (2013). Role of stem-cell-derived microvesicles in the paracrine action of stem cells. Biochem. Soc. Trans. 41, 283–287. doi: 10.1042/BST20120192

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, J., and Chopp, M. (2018). Exosome therapy for stroke. Stroke 49, 1083–1090. doi: 10.1161/STROKEAHA.117.018292

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, Y., Li, J., Ma, B., Li, N., Wang, S., Sun, Z., et al. (2020). MSC-derived exosomes promote recovery from traumatic brain injury via microglia/macrophages in rat. Aging (Albany NY) 12, 18274–18296. doi: 10.18632/aging.103692

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, J., Li, Y., Wang, L., Zhang, Z., Lu, D., Lu, M., et al. (2001). Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke 32, 1005–1011. doi: 10.1161/01.STR.32.4.1005

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, C. C., Liu, L., Ma, F., Wong, C. W., Guo, X. E., Chacko, J. V., et al. (2016). Elucidation of exosome migration across the blood-brain barrier model in vitro. Cell. Mol. Bioeng. 9, 509–529. doi: 10.1007/s12195-016-0458-3

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, P., Price, C., Li, Z. J., Li, Y. Y., Cao, D. L., Wiley, A., et al. (2013). miR-9 is an essential oncogenic microRNA specifically overexpressed in rearranged leukemia. Proc. Natl. Acad. Sci. USA 110, 11511–11516. doi: 10.1073/pnas.1310144110

PubMed Abstract | Crossref Full Text | Google Scholar

Chopp, M., and Li, Y. (2002). Treatment of neural injury with marrow stromal cells. Lancet Neurol. 1, 92–100. doi: 10.1016/S1474-4422(02)00040-6

Crossref Full Text | Google Scholar

Chopp, M., and Zhang, Z. G. (2015). Emerging potential of exosomes and noncoding microRNAs for the treatment of neurological injury/diseases. Expert Opin. Emerg. Drugs 20, 523–526. doi: 10.1517/14728214.2015.1061993

PubMed Abstract | Crossref Full Text | Google Scholar

Cox, C. S. Jr., Baumgartner, J. E., Harting, M. T., Worth, L. L., Walker, P. A., Shah, S. K., et al. (2011). Autologous bone marrow mononuclear cell therapy for severe traumatic brain injury in children. Neurosurgery 68, 588–600. doi: 10.1227/NEU.0b013e318207734c

PubMed Abstract | Crossref Full Text | Google Scholar

Deng, Y., Chen, D., Gao, F., Lv, H., Zhang, G., Sun, X., et al. (2019). Exosomes derived from microRNA-138-5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2. J. Biol. Eng. 13:71. doi: 10.1186/s13036-019-0193-0

PubMed Abstract | Crossref Full Text | Google Scholar

Di Pietro, V., Ragusa, M., Davies, D., Su, Z. J., Hazeldine, J., Lazzarino, G., et al. (2017). MicroRNAs as novel biomarkers for the diagnosis and prognosis of mild and severe traumatic brain injury. J. Neurotrauma 34, 1948–1956. doi: 10.1089/neu.2016.4857

PubMed Abstract | Crossref Full Text | Google Scholar

Doeppner, T. R., Herz, J., Gorgens, A., Schlechter, J., Ludwig, A. K., Radtke, S., et al. (2015). Extracellular vesicles improve post-stroke neuroregeneration and prevent postischemic immunosuppression. Stem Cells Transl. Med. 4, 1131–1143. doi: 10.5966/sctm.2015-0078

PubMed Abstract | Crossref Full Text | Google Scholar

Duan, S., Wang, F., Cao, J., and Wang, C. (2020). Exosomes derived from MicroRNA-146a-5p-enriched bone marrow mesenchymal stem cells alleviate intracerebral hemorrhage by inhibiting neuronal apoptosis and microglial M1 polarization. Drug Des. Devel. Ther. 14, 3143–3158. doi: 10.2147/DDDT.S255828

PubMed Abstract | Crossref Full Text | Google Scholar

Dumbrava, D. A., Surugiu, R., Borger, V., Ruscu, M., Tertel, T., Giebel, B., et al. (2022). Mesenchymal stromal cell-derived small extracellular vesicles promote neurological recovery and brain remodeling after distal middle cerebral artery occlusion in aged rats. Geroscience 44, 293–310. doi: 10.1007/s11357-021-00483-2

PubMed Abstract | Crossref Full Text | Google Scholar

El Andaloussi, S., Mager, I., Breakefield, X. O., and Wood, M. J. (2013). Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov. 12, 347–357. doi: 10.1038/nrd3978

Crossref Full Text | Google Scholar

Fruhbeis, C., Frohlich, D., Kuo, W. P., Amphornrat, J., Thilemann, S., Saab, A. S., et al. (2013a). Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 11:e1001604. doi: 10.1371/journal.pbio.1001604

PubMed Abstract | Crossref Full Text | Google Scholar

Fruhbeis, C., Frohlich, D., Kuo, W. P., and Kramer-Albers, E. M. (2013b). Extracellular vesicles as mediators of neuron-glia communication. Front. Cell. Neurosci. 7:182. doi: 10.3389/fncel.2013.00182

Crossref Full Text | Google Scholar

Gao, W., Li, F., Liu, L., Xu, X., Zhang, B., Wu, Y., et al. (2018). Endothelial colony-forming cell-derived exosomes restore blood-brain barrier continuity in mice subjected to traumatic brain injury. Exp. Neurol. 307, 99–108. doi: 10.1016/j.expneurol.2018.06.001

PubMed Abstract | Crossref Full Text | Google Scholar

Gao, X., Xiong, Y., Li, Q., Han, M., Shan, D., Yang, G., et al. (2020). Extracellular vesicle-mediated transfer of miR-21-5p from mesenchymal stromal cells to neurons alleviates early brain injury to improve cognitive function via the PTEN/Akt pathway after subarachnoid hemorrhage. Cell Death Dis. 11:363. doi: 10.1038/s41419-020-2530-0

PubMed Abstract | Crossref Full Text | Google Scholar

Ge, X., Guo, M., Hu, T., Li, W., Huang, S., Yin, Z., et al. (2020). Increased microglial exosomal miR-124-3p alleviates neurodegeneration and improves cognitive outcome after rmTBI. Mol. Ther. 28, 503–522. doi: 10.1016/j.ymthe.2019.11.017

PubMed Abstract | Crossref Full Text | Google Scholar

Gibbings, D. J., Ciaudo, C., Erhardt, M., and Voinnet, O. (2009). Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat. Cell Biol. 11, 1143–1149. doi: 10.1038/ncb1929

PubMed Abstract | Crossref Full Text | Google Scholar

Giovannelli, L., Bari, E., Jommi, C., Tartara, F., Armocida, D., Garbossa, D., et al. (2023). Mesenchymal stem cell secretome and extracellular vesicles for neurodegenerative diseases: risk-benefit profile and next steps for the market access. Bioact Mater. 29, 16–35. doi: 10.1016/j.bioactmat.2023.06.013

PubMed Abstract | Crossref Full Text | Google Scholar

Go, V., Bowley, B. G. E., Pessina, M. A., Zhang, Z. G., Chopp, M., Finklestein, S. P., et al. (2020). Extracellular vesicles from mesenchymal stem cells reduce microglial-mediated neuroinflammation after cortical injury in aged Rhesus monkeys. Geroscience 42, 1–17. doi: 10.1007/s11357-019-00115-w

PubMed Abstract | Crossref Full Text | Google Scholar

Go, V., Sarikaya, D., Zhou, Y., Bowley, B. G. E., Pessina, M. A., Rosene, D. L., et al. (2021). Extracellular vesicles derived from bone marrow mesenchymal stem cells enhance myelin maintenance after cortical injury in aged rhesus monkeys. Exp. Neurol. 337:113540. doi: 10.1016/j.expneurol.2020.113540

PubMed Abstract | Crossref Full Text | Google Scholar

Hacke, W., Kaste, M., Bluhmki, E., Brozman, M., Davalos, A., Guidetti, D., et al. (2008). Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N. Engl. J. Med. 359, 1317–1329. doi: 10.1056/NEJMoa0804656

Crossref Full Text | Google Scholar

Hering, C., and Shetty, A. K. (2023). Extracellular vesicles derived from neural stem cells, astrocytes, and microglia as therapeutics for easing TBI-induced brain dysfunction. Stem Cells Transl. Med. 12, 140–153. doi: 10.1093/stcltm/szad004

PubMed Abstract | Crossref Full Text | Google Scholar

Huang, L. Y., Song, J. X., Cai, H., Wang, P. P., Yin, Q. L., Zhang, Y. D., et al. (2022). Healthy serum-derived exosomes improve neurological outcomes and protect blood-brain barrier by inhibiting endothelial cell apoptosis and reversing autophagy-mediated tight junction protein reduction in rat stroke model. Front. Cell. Neurosci. 16:841544. doi: 10.3389/fncel.2022.841544

PubMed Abstract | Crossref Full Text | Google Scholar

Hyder, A. A., Wunderlich, C. A., Puvanachandra, P., Gururaj, G., and Kobusingye, O. C. (2007). The impact of traumatic brain injuries: a global perspective. NeuroRehabilitation 22, 341–353. doi: 10.3233/NRE-2007-22502

PubMed Abstract | Crossref Full Text | Google Scholar

Jayaraman, M., Radhakrishnan, R., Mathews, C. A., Yan, M., Husain, S., Moxley, K. M., et al. (2017). Identification of novel diagnostic and prognostic miRNA signatures in endometrial cancer. Genes Cancer 8, 566–576. doi: 10.18632/genesandcancer.144

PubMed Abstract | Crossref Full Text | Google Scholar

Jiang, M., Wang, H., Jin, M., Yang, X., Ji, H., Jiang, Y., et al. (2018). Exosomes from MiR-30d-5p-ADSCs reverse acute ischemic stroke-induced, autophagy-mediated brain injury by promoting M2 microglial/macrophage polarization. Cell. Physiol. Biochem. 47, 864–878. doi: 10.1159/000490078

PubMed Abstract | Crossref Full Text | Google Scholar

Johnson, W. D., and Griswold, D. P. (2017). Traumatic brain injury: a global challenge. Lancet Neurol. 16, 949–950. doi: 10.1016/S1474-4422(17)30362-9

Crossref Full Text | Google Scholar

Kalluri, R., and LeBleu, V. S. (2020). The biology, function, and biomedical applications of exosomes. Science 2, 173–174. doi: 10.1126/science.aau6977

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, D. K., Nishida, H., An, S. Y., Shetty, A. K., Bartosh, T. J., and Prockop, D. J. (2016). Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc. Natl. Acad. Sci. USA 113, 170–175. doi: 10.1073/pnas.1522297113

PubMed Abstract | Crossref Full Text | Google Scholar

Kramer-Albers, E. M., Bretz, N., Tenzer, S., Winterstein, C., Mobius, W., Berger, H., et al. (2007). Oligodendrocytes secrete exosomes containing major myelin and stress-protective proteins: trophic support for axons? Proteomics Clin. Appl. 1, 1446–1461. doi: 10.1002/prca.200700522

PubMed Abstract | Crossref Full Text | Google Scholar

Kuriakose, D., and Xiao, Z. (2020). Pathophysiology and treatment of stroke: present status and future perspectives. Int. J. Mol. Sci. 21, 1–102. doi: 10.3390/ijms21207609

PubMed Abstract | Crossref Full Text | Google Scholar

Lackland, D. T., Roccella, E. J., Deutsch, A. F., Fornage, M., George, M. G., Howard, G., et al. (2014). Factors influencing the decline in stroke mortality: a statement from the American Heart Association/American Stroke Association. Stroke 45, 315–353. doi: 10.1161/01.str.0000437068.30550.cf

PubMed Abstract | Crossref Full Text | Google Scholar

Lafourcade, C., Ramirez, J. P., Luarte, A., Fernandez, A., and Wyneken, U. (2016). MiRNAs in astrocyte-derived exosomes as possible mediators of neuronal plasticity. J. Exp. Neurosci. 10, 1–9. doi: 10.4137/JEN.S39916

PubMed Abstract | Crossref Full Text | Google Scholar

Lai, C. P., and Breakefield, X. O. (2012). Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front. Physiol. 3:228. doi: 10.3389/fphys.2012.00228

Crossref Full Text | Google Scholar

Lai, R. C., Chen, T. S., and Lim, S. K. (2011). Mesenchymal stem cell exosome: a novel stem cell-based therapy for cardiovascular disease. Regen. Med. 6, 481–492. doi: 10.2217/rme.11.35

PubMed Abstract | Crossref Full Text | Google Scholar

Lai, N. S., Wu, D. G., Liang, T. Y., Pan, P. J., Yuan, G. Q., Li, X., et al. (2020). Systemic exosomal miR-193b-3p delivery attenuates neuroinflammation in early brain injury after subarachnoid hemorrhage in mice. J. Neuroinflammation 17:74. doi: 10.1186/s12974-020-01745-0

PubMed Abstract | Crossref Full Text | Google Scholar

Latchana, N., Abrams, Z. B., Howard, J. H., Regan, K., Jacob, N., Fadda, P., et al. (2017). Plasma microRNA levels following resection of metastatic melanoma. Bioinform. Biol. Insights 11:117793221769483. doi: 10.1177/1177932217694837

Crossref Full Text | Google Scholar

Lener, T., Gimona, M., Aigner, L., Borger, V., Buzas, E., Camussi, G., et al. (2015). Applying extracellular vesicles based therapeutics in clinical trials – an ISEV position paper. J. Extracell. Vesicles 4:30087. doi: 10.3402/jev.v4.30087

PubMed Abstract | Crossref Full Text | Google Scholar

Li, D., Huang, S., Yin, Z., Zhu, J., Ge, X., Han, Z., et al. (2019). Increases in miR-124-3p in microglial exosomes confer neuroprotective effects by targeting FIP200-mediated neuronal autophagy following traumatic brain injury. Neurochem. Res. 44, 1903–1923. doi: 10.1007/s11064-019-02825-1

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, Y. Y., Li, Y., Wang, L., Zhao, Y., Yuan, R., Yang, M. M., et al. (2023). Mesenchymal stem cell-derived exosomes regulate microglia phenotypes: a promising treatment for acute central nervous system injury. Neural Regen. Res. 18, 1657–1665. doi: 10.4103/1673-5374.363819

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, X., Zhang, L., Cao, Y., Jia, H., Li, X., Li, F., et al. (2022). Neuroinflammation of traumatic brain injury: roles of extracellular vesicles. Front. Immunol. 13:1088827. doi: 10.3389/fimmu.2022.1088827

Crossref Full Text | Google Scholar

Liu, X., Zhang, M., Liu, H., Zhu, R., He, H., Zhou, Y., et al. (2021). Bone marrow mesenchymal stem cell-derived exosomes attenuate cerebral ischemia-reperfusion injury-induced neuroinflammation and pyroptosis by modulating microglia M1/M2 phenotypes. Exp. Neurol. 341:113700. doi: 10.1016/j.expneurol.2021.113700

PubMed Abstract | Crossref Full Text | Google Scholar

Macfarlane, L. A., and Murphy, P. R. (2010). MicroRNA: biogenesis, function and role in cancer. Curr. Genomics 11, 537–561. doi: 10.2174/138920210793175895

PubMed Abstract | Crossref Full Text | Google Scholar

Mateescu, B., Kowal, E. J., van Balkom, B. W., Bartel, S., Bhattacharyya, S. N., Buzas, E. I., et al. (2017). Obstacles and opportunities in the functional analysis of extracellular vesicle RNA – an ISEV position paper. J. Extracell. Vesicles 6:1286095. doi: 10.1080/20013078.2017.1286095

PubMed Abstract | Crossref Full Text | Google Scholar

Mavroudis, I., Balmus, I. M., Ciobica, A., Nicoara, M. N., Luca, A. C., and Palade, D. O. (2023). The role of microglial exosomes and miR-124-3p in neuroinflammation and neuronal repair after traumatic brain injury. Life (Basel) 13, 2–188. doi: 10.3390/life13091924

Crossref Full Text | Google Scholar

Moore, T. L., Bowley, B. G. E., Pessina, M. A., Calderazzo, S. M., Medalla, M., Go, V., et al. (2019). Mesenchymal derived exosomes enhance recovery of motor function in a monkey model of cortical injury. Restor. Neurol. Neurosci. 37, 347–362. doi: 10.3233/RNN-190910

PubMed Abstract | Crossref Full Text | Google Scholar

Nasirishargh, A., Kumar, P., Ramasubramanian, L., Clark, K., Hao, D., Lazar, S. V., et al. (2021). Exosomal microRNAs from mesenchymal stem/stromal cells: biology and applications in neuroprotection. World J. Stem Cells 13, 776–794. doi: 10.4252/wjsc.v13.i7.776

PubMed Abstract | Crossref Full Text | Google Scholar

National Institute of Neurological Disorders and Stroke rt, PA Stroke Study Group (1995). Tissue plasminogen activator for acute ischemic stroke. N. Engl. J. Med. 333, 1581–1587.

Google Scholar

Nichols, J. E., Niles, J. A., DeWitt, D., Prough, D., Parsley, M., Vega, S., et al. (2013). Neurogenic and neuro-protective potential of a novel subpopulation of peripheral blood-derived CD133+ ABCG2+CXCR4+ mesenchymal stem cells: development of autologous cell-based therapeutics for traumatic brain injury. Stem Cell Res Ther 4:3. doi: 10.1186/scrt151

PubMed Abstract | Crossref Full Text | Google Scholar

O'Brien, J., Hayder, H., Zayed, Y., and Peng, C. (2018). Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front. Endocrinol. (Lausanne) 9:402. doi: 10.3389/fendo.2018.00402

PubMed Abstract | Crossref Full Text | Google Scholar

Otero-Ortega, L., Gomez de Frutos, M. C., Laso-Garcia, F., Rodriguez-Frutos, B., Medina-Gutierrez, E., Lopez, J. A., et al. (2018). Exosomes promote restoration after an experimental animal model of intracerebral hemorrhage. J. Cereb. Blood Flow Metab. 38, 767–779. doi: 10.1177/0271678X17708917

PubMed Abstract | Crossref Full Text | Google Scholar

Otero-Ortega, L., Laso-Garcia, F., Gomez-de Frutos, M. D., Rodriguez-Frutos, B., Pascual-Guerra, J., Fuentes, B., et al. (2017). White matter repair after extracellular vesicles administration in an experimental animal model of subcortical stroke. Sci. Rep. 7:44433. doi: 10.1038/srep44433

PubMed Abstract | Crossref Full Text | Google Scholar

Pan, Y. B., Sun, Z. L., and Feng, D. F. (2017). The role of MicroRNA in traumatic brain injury. Neuroscience 367, 189–199. doi: 10.1016/j.neuroscience.2017.10.046

Crossref Full Text | Google Scholar

Panaro, M. A., Benameur, T., and Porro, C. (2020). Extracellular vesicles miRNA cargo for microglia polarization in traumatic brain injury. Biomol. Ther. 10, 5–513. doi: 10.3390/biom10060901

PubMed Abstract | Crossref Full Text | Google Scholar

Phinney, D. G., and Prockop, D. J. (2007). Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair—current views. Stem Cells 25, 2896–2902. doi: 10.1634/stemcells.2007-0637

PubMed Abstract | Crossref Full Text | Google Scholar

Pischiutta, F., Caruso, E., Cavaleiro, H., Salgado, A. J., Loane, D. J., and Zanier, E. R. (2022). Mesenchymal stromal cell secretome for traumatic brain injury: focus on immunomodulatory action. Exp. Neurol. 357:114199. doi: 10.1016/j.expneurol.2022.114199

PubMed Abstract | Crossref Full Text | Google Scholar

Rak, J. (2013). Extracellular vesicles – biomarkers and effectors of the cellular interactome in cancer. Front. Pharmacol. 4:21. doi: 10.3389/fphar.2013.00021

Crossref Full Text | Google Scholar

Redell, J. B., Moore, A. N., Ward, N. H. 3rd, Hergenroeder, G. W., and Dash, P. K. (2010). Human traumatic brain injury alters plasma microRNA levels. J. Neurotrauma 27, 2147–2156. doi: 10.1089/neu.2010.1481

PubMed Abstract | Crossref Full Text | Google Scholar

Schneider, A., and Simons, M. (2013). Exosomes: vesicular carriers for intercellular communication in neurodegenerative disorders. Cell Tissue Res. 352, 33–47. doi: 10.1007/s00441-012-1428-2

PubMed Abstract | Crossref Full Text | Google Scholar

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

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, P., Liu, D. Z., Jickling, G. C., Sharp, F. R., and Yin, K. J. (2018). MicroRNA-based therapeutics in central nervous system injuries. J. Cereb. Blood Flow Metab. 38, 1125–1148. doi: 10.1177/0271678X18773871

PubMed Abstract | Crossref Full Text | Google Scholar

Thery, C., Regnault, A., Garin, J., Wolfers, J., Zitvogel, L., Ricciardi-Castagnoli, P., et al. (1999). Molecular characterization of dendritic cell-derived exosomes. Selective accumulation of the heat shock protein hsc73. J. Cell Biol. 147, 599–610. doi: 10.1083/jcb.147.3.599

PubMed Abstract | Crossref Full Text | Google Scholar

Thery, C., Zitvogel, L., and Amigorena, S. (2002). Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–579. doi: 10.1038/nri855

Crossref Full Text | Google Scholar

van Niel, G., Porto-Carreiro, I., Simoes, S., and Raposo, G. (2006). Exosomes: a common pathway for a specialized function. J. Biochem. 140, 13–21. doi: 10.1093/jb/mvj128

Crossref Full Text | Google Scholar

Wang, J., Chen, S., Zhang, W., Chen, Y., and Bihl, J. C. (2020). Exosomes from miRNA-126-modified endothelial progenitor cells alleviate brain injury and promote functional recovery after stroke. CNS Neurosci. Ther. 26, 1255–1265. doi: 10.1111/cns.13455

PubMed Abstract | Crossref Full Text | Google Scholar

Webb, R. L., Kaiser, E. E., Jurgielewicz, B. J., Spellicy, S., Scoville, S. L., Thompson, T. A., et al. (2018). Human neural stem cell extracellular vesicles improve recovery in a porcine model of ischemic stroke. Stroke 49, 1248–1256. doi: 10.1161/STROKEAHA.117.020353

PubMed Abstract | Crossref Full Text | Google Scholar

Wen, M. M. (2016). Getting miRNA therapeutics into the target cells for neurodegenerative diseases: a Mini-review. Front. Mol. Neurosci. 9:129. doi: 10.3389/fnmol.2016.00129

PubMed Abstract | Crossref Full Text | Google Scholar

Williams, A. M., Higgins, G. A., Bhatti, U. F., Biesterveld, B. E., Dekker, S. E., Kathawate, R. G., et al. (2020a). Early treatment with exosomes following traumatic brain injury and hemorrhagic shock in a swine model promotes transcriptional changes associated with neuroprotection. J. Trauma Acute Care Surg. 89, 536–543. doi: 10.1097/TA.0000000000002815

Crossref Full Text | Google Scholar

Williams, A. M., Wu, Z., Bhatti, U. F., Biesterveld, B. E., Kemp, M. T., Wakam, G. K., et al. (2020b). Early single-dose exosome treatment improves neurologic outcomes in a 7-day swine model of traumatic brain injury and hemorrhagic shock. J. Trauma Acute Care Surg. 89, 388–396. doi: 10.1097/TA.0000000000002698

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, K., Yang, Y., Zhong, Y., Ammar, H. M., Zhang, P., Guo, R., et al. (2016). The effects of microvesicles on endothelial progenitor cells are compromised in type 2 diabetic patients via downregulation of the miR-126/VEGFR2 pathway. Am. J. Physiol. Endocrinol. Metab. 310, E828–E837. doi: 10.1152/ajpendo.00056.2016

PubMed Abstract | Crossref Full Text | Google Scholar

Xia, X. H., Wang, Y., Qin, Y., Zhao, S., and Zheng, J. L. C. (2022). Exosome: a novel neurotransmission modulator or non-canonical neurotransmitter? Ageing Res. Rev. 74:101558. doi: 10.1016/j.arr.2021.101558

Crossref Full Text | Google Scholar

Xiao, R., Wang, Q., Peng, J., Yu, Z., Zhang, J., and Xia, Y. (2023). BMSC-derived exosomal Egr2 ameliorates ischemic stroke by directly upregulating SIRT6 to suppress notch signaling. Mol. Neurobiol. 60, 1–17. doi: 10.1007/s12035-022-03037-5

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, H., Katakowski, M., Wang, F., Qian, J. Y., Liu, X. S., Ali, M. M., et al. (2017a). MicroRNA cluster miR-17-92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke 48, 747–753. doi: 10.1161/STROKEAHA.116.015204

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, H., Li, Y., and Chopp, M. (2014). Exosomes/miRNAs as mediating cell-based therapy of stroke. Front. Cell. Neurosci. 8:377. doi: 10.3389/fncel.2014.00377

Crossref Full Text | Google Scholar

Xin, H., Li, Y., Cui, Y., Yang, J. J., Zhang, Z. G., and Chopp, M. (2013a). Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J. Cereb. Blood Flow Metab. 33, 1711–1715. doi: 10.1038/jcbfm.2013.152

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, H., Li, Y., Liu, Z., Wang, X., Shang, X., Cui, Y., et al. (2013b). MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem Cells 31, 2737–2746. doi: 10.1002/stem.1409

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, H., Liu, Z., Buller, B., Li, Y., Golembieski, W., Gan, X., et al. (2021). MiR-17-92 enriched exosomes derived from multipotent mesenchymal stromal cells enhance axon-myelin remodeling and motor electrophysiological recovery after stroke. J. Cereb. Blood Flow Metab. 41, 1131–1144. doi: 10.1177/0271678X20950489

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, H., Wang, F., Li, Y., Lu, Q. E., Cheung, W. L., Zhang, Y., et al. (2017b). Secondary release of exosomes from astrocytes contributes to the increase in neural plasticity and improvement of functional recovery after stroke in rats treated with exosomes harvested from microRNA 133b-overexpressing multipotent mesenchymal stromal cells. Cell Transplant. 26, 243–257. doi: 10.3727/096368916X693031

PubMed Abstract | Crossref Full Text | Google Scholar

Xiong, Y., Mahmood, A., and Chopp, M. (2017). Emerging potential of exosomes for treatment of traumatic brain injury. Neural Regen. Res. 12, 19–22. doi: 10.4103/1673-5374.198966

PubMed Abstract | Crossref Full Text | Google Scholar

Xiong, Y., Mahmood, A., and Chopp, M. (2018). Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin. J. Traumatol. 21, 137–151. doi: 10.1016/j.cjtee.2018.02.003

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, B., Zhang, Y., Du, X. F., Li, J., Zi, H. X., Bu, J. W., et al. (2017). Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res. 27, 882–897. doi: 10.1038/cr.2017.62

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, X., Zhang, H., Li, J., Chen, Y., Zhong, W., Chen, Y., et al. (2023). Combination of EPC-EXs and NPC-EXs with miR-126 and miR-210 overexpression produces better therapeutic effects on ischemic stroke by protecting neurons through the Nox2/ROS and BDNF/TrkB pathways. Exp. Neurol. 359:114235. doi: 10.1016/j.expneurol.2022.114235

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, Y., Ye, Y., Kong, C., Su, X., Zhang, X., Bai, W., et al. (2019). MiR-124 enriched exosomes promoted the M2 polarization of microglia and enhanced hippocampus neurogenesis after traumatic brain injury by inhibiting TLR4 pathway. Neurochem. Res. 44, 811–828. doi: 10.1007/s11064-018-02714-z

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, Y., Ye, Y., Su, X., He, J., Bai, W., and He, X. (2017). MSCs-derived exosomes and neuroinflammation, neurogenesis and therapy of traumatic brain injury. Front. Cell. Neurosci. 11:55. doi: 10.3389/fncel.2017.00055

Crossref Full Text | Google Scholar

Yang, J., Zhang, X., Chen, X., Wang, L., and Yang, G. (2017). Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia. Mol. Ther. Nucleic Acids 7, 278–287. doi: 10.1016/j.omtn.2017.04.010

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Z. G., Buller, B., and Chopp, M. (2019). Exosomes – beyond stem cells for restorative therapy in stroke and neurological injury. Nat. Rev. Neurol. 15, 193–203. doi: 10.1038/s41582-018-0126-4

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Z. G., and Chopp, M. (2009). Neurorestorative therapies for stroke: underlying mechanisms and translation to the clinic. Lancet Neurol. 8, 491–500. doi: 10.1016/S1474-4422(09)70061-4

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Chopp, M., Liu, X. S., Katakowski, M., Wang, X., Tian, X., et al. (2017a). Exosomes derived from mesenchymal stromal cells promote axonal growth of cortical neurons. Mol. Neurobiol. 54, 2659–2673. doi: 10.1007/s12035-016-9851-0

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Chopp, M., Meng, Y., Katakowski, M., Xin, H., Mahmood, A., et al. (2015). Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J. Neurosurg. 122, 856–867. doi: 10.3171/2014.11.JNS14770

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Chopp, M., Zhang, Z. G., Katakowski, M., Xin, H., Qu, C., et al. (2017b). Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem. Int. 111, 69–81. doi: 10.1016/j.neuint.2016.08.003

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, W., Hong, J., Zhang, H., Zheng, W., and Yang, Y. (2021). Astrocyte-derived exosomes protect hippocampal neurons after traumatic brain injury by suppressing mitochondrial oxidative stress and apoptosis. Aging (Albany NY) 13, 21642–21658. doi: 10.18632/aging.203508

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, R., Mao, W., Niu, L., Bao, W., Wang, Y., Wang, Y., et al. (2023). NSC-derived exosomes enhance therapeutic effects of NSC transplantation on cerebral ischemia in mice. eLife 12, 3-248, 254-279–4-403. doi: 10.7554/eLife.84493

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, H., Mao, F., Shen, T., Luo, Q., Ding, Z., Qian, L., et al. (2017). Plasma miR-145, miR-20a, miR-21 and miR-223 as novel biomarkers for screening early-stage non-small cell lung cancer. Oncol. Lett. 13, 669–676. doi: 10.3892/ol.2016.5462

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, L., Mao, L., and Wang, H. (2022). The neuroprotection effects of exosome in central nervous system injuries: a new target for therapeutic intervention. Mol. Neurobiol. 59, 7152–7169. doi: 10.1007/s12035-022-03028-6

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, R. L., Pan, W. L., Zhang, X. M., Liu, X. S., Landschoot-Ward, J., Li, C., et al. (2017). Cerebral endothelial derived exosomes abolish cognitive impairment induced by ablation of dicer in adult neural progenitor cells. Stroke 48, 3–323. doi: 10.1161/str.48.suppl_1.wmp48

Crossref Full Text | Google Scholar

Zhang, Y., Zhang, Y., Chopp, M., Pang, H., Chen, L., Zhang, Z. G., et al. (2023). Therapeutic role of microRNAs of small extracellular vesicles from human mesenchymal stromal/stem cells in treatment of experimental traumatic brain injury. J. Neurotrauma 40, 758–771. doi: 10.1089/neu.2022.0296

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Zhang, Y., Chopp, M., Pang, H., Zhang, Z. G., Mahmood, A., et al. (2021). MiR-17-92 cluster-enriched exosomes derived from human bone marrow mesenchymal stromal cells improve tissue and functional recovery in rats after traumatic brain injury. J. Neurotrauma 38, 1535–1550. doi: 10.1089/neu.2020.7575

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Zhang, Y., Chopp, M., Zhang, Z. G., Mahmood, A., and Xiong, Y. (2020). Mesenchymal stem cell-derived exosomes improve functional recovery in rats after traumatic brain injury: a dose-response and therapeutic window study. Neurorehabil. Neural Repair 34, 616–626. doi: 10.1177/1545968320926164

PubMed Abstract | Crossref Full Text | Google Scholar

Zhou, H., Zhou, J., Teng, H., Yang, H., Qiu, J., and Li, X. (2022). MiR-145 enriched exosomes derived from bone marrow-derived mesenchymal stem cells protects against cerebral ischemia-reperfusion injury through downregulation of FOXO1. Biochem. Biophys. Res. Commun. 632, 92–99. doi: 10.1016/j.bbrc.2022.09.089

PubMed Abstract | Crossref Full Text | Google Scholar

Zhuang, Z., Liu, M., Dai, Z., Luo, J., Zhang, B., Yu, H., et al. (2023). Bone marrow stromal cells-derived exosomes reduce neurological damage in traumatic brain injury through the miR-124-3p/p38 MAPK/GLT-1 axis. Exp. Neurol. 365:114408. doi: 10.1016/j.expneurol.2023.114408

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: exosome, stroke, traumatic brain injury (TBI), miRNA, engineered exosome

Citation: Chen L, Xiong Y, Chopp M and Zhang Y (2024) Engineered exosomes enriched with select microRNAs amplify their therapeutic efficacy for traumatic brain injury and stroke. Front. Cell. Neurosci. 18:1376601. doi: 10.3389/fncel.2024.1376601

Received: 25 January 2024; Accepted: 29 February 2024;
Published: 19 March 2024.

Edited by:

Dirk M. Hermann, University of Duisburg-Essen, Germany

Reviewed by:

Mingzi Zhang, University of Southern California, United States

Copyright © 2024 Chen, Xiong, Chopp and Zhang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yanlu Zhang, yzhang2@hfhs.org

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.