11

Treating Cerebral Ischemia: Novel Therapeutic Strategies from Experimental Stroke Research

Xuan Zheng Matteo Haupt Mathias Bähr Lars Tatenhorst Thorsten R. Doeppner

Department of Neurology, University of Göttingen Medical School, Göttingen, Germany

Abstract: Although systemic thrombolysis and endovascular treatment have revolutionized modern stroke treatment, the majority of patients do not qualify for either treatment paradigm. Hence, novel adjuvant therapeutic strategies are required. This chapter provides an overview of our current understanding of novel therapeutic strategies in preclinical stroke models. The chapter is organized in three major parts to cover the acute, subacute, and chronic phases of ischemic stroke. The potential of various pharmacological agents, stem cells, microRNAs, and extracellular vesicles as therapeutic avenues along with the progress and challenges are discussed.

Keywords: cerebral ischemia; extracellular vesicles; neuroprotection; neurodegeneration; stroke

Author for correspondence: Thorsten R. Doeppner, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany. Email: thorsten.doeppner@med.uni-goettingen.de

Doi: https://doi.org/10.36255/exonpublications.cerebralischemia.2021.therapy

In: Cerebral Ischemia. Pluta R (Editor). Exon Publications, Brisbane, Australia. ISBN: 978-0-6450017-9-2; Doi: https://doi.org/10.36255/exonpublications.cerebralischemia.2021

Copyright: The Authors.

License: This open access article is licenced under Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0) https://creativecommons.org/licenses/by-nc/4.0/

INTRODUCTION

The temporal patterns of stroke are divided into acute, subacute and chronic stages of the disease (1). The diagnostic procedures for patients with clinical signs of an acute ischemic stroke include brain imaging techniques such as computed tomography (CT) and magnetic resonance imaging (MRI) (2). Beside these well-established imaging tools, a variety of novel stroke biomarkers including proteins, lipids, RNAs, and metabolites are subject to current research (3, 4). However, none of them has been translated into routine clinical settings, yet. Stroke is an emergency, and time is a crucial factor for not only diagnostic but also therapeutic procedures. Causal treatments of acute ischemic stroke include systemic thrombolysis and mechanic thrombectomy (5), which are reserved for a minority of patients because of contraindications and narrow time windows. In addition, revascularization itself does not halt the stroke-induced activation of proinjurious signaling cascades within the ischemic tissue. The availability of novel adjuvant neuroprotective tools may therefore lead to a paradigm shift in stroke therapy. Numerous preclinical studies with potential therapeutic agents that modulate the pathophysiological process in the acute, subacute, and chronic phases of ischemic stroke have been conducted. However, despite extensive research, translation from preclinical findings to clinical routine has not yet been successful.

ACUTE PHASE OF STROKE

For the acute phase of stroke, research activities have predominantly focused on neuroprotection. Many pharmacologically active drugs have been found to be neuroprotective in preclinical stroke settings (6). Such pharmacological interventions target various aspects of the complex pathophysiology of stroke, among which are excitotoxicity, oxidative stress, calcium influx, programmed cell death, immune system response, and blood-brain barrier (BBB) disruption (7). Although translation of promising preclinical data into the clinic has failed thus far (8), promising research activities continue to occur worldwide. Some of these novel treatment strategies are discussed in the following paragraphs (Figure 1).

Fig 1

Figure 1. Main targets in acute phase of ischemic stroke by neuroprotective agents. Preclinical neuroprotective candidates mainly target one or more of various key players during the acute stage of ischemic stroke. These targets include oxidative stress, excitotoxicity, calcium influx, immune response and programmed cell death, all of which are tightly interconnected. Examples of particular downstream mechanisms are shown. Abbreviations: nNOS, neuronal nitric oxide synthase; NMDAR, N-Methyl-D-aspartic acid receptor.

Glutamate-mediated excitotoxicity, which involves N-Methyl-D-Aspartat receptors (NMDAR), has been considered as one of the most important cell death mechanisms in the pathogenesis of stroke (9). Several drugs, particularly inhibiting NMDAR, were found to be neuroprotective in preclinical studies (10). Beside directly inhibiting NMDAR, some studies focus on analyzing downstream targets (11). For instance, the PSD95 inhibitor Tat-NR2B9c (NA-1) has shown neuroprotective effects by decreasing the ability of NMDAR to trigger downstream excitotoxicity via neuronal nitric oxide synthase (nNOS) activation (11).

Ischemia-induced oxidative stress leads to vasodilatation and altered vascular reactivity which in turn promote BBB breakdown and severe tissue injury (12). Oxidative stress includes the generation of superoxides and nitric oxides, which are both linked to excitotoxicity (13, 14). Free radical scavengers like edaravone, uric acid, and citicoline have been identified as potential neuroprotective agents (15). Excessive Ca2+-influx leading to toxic intracellular levels is a major factor for triggering detrimental ischemic signal cascades, such as the activation of proapoptotic enzymes and synthesis of free radicals (7). Thus, pharmacological inhibition of Ca2+ overload is a major field in neuroprotective stroke research. There are two major strategies: blocking of calcium channels, and modulation of calcium-related signaling molecules. Calcium channel blockers (CCB) such as lercanidipine and cilnidipine lead to neuroprotection by attenuating oxidative stress, inflammation and apoptosis (16, 17). Furthermore, intraarterial delivery of Verapamil, a routinely used CCB in the treatment of cardiac diseases, leads to neuroprotection in experimental models of stroke (18). A promising candidate for regulating the calcium-related downstream molecules is paeoniflorin. Paeoniflorin has been shown to decrease neurological deficit scores and infarct volumes in rats by regulating the Ca2+/calmodulin-dependent protein kinase II/response element-binding signaling pathway (19).

Ischemia-induced changes in cellular homeostasis trigger programmed cell death such as apoptosis, necroptosis, and autophagy. The cell death mechanism of each single cell is mainly determined by the extracellular milieu, cell type, cell age, and location in the brain (20). It is worth noting that in the last decades the view on programmed cell death has changed, since additional forms and hybrid forms were discovered (21). Modulating these ischemia-induced cell death signal cascades is one of the major experimental approaches to reduce neuronal cell loss after ischemic stroke. Caspase inhibitors, for example the caspase-3 inhibitor z-DEVD-fm, effectively inhibit apoptosis and lead to neuroprotection in rodent models (22, 23). On the contrary, necroptosis has been found to have caspase-independent regulation pathways (24). Pharmacological inhibition of the latter using receptor-interacting serine/threonine-protein kinase (RIP) inhibitors lead to a reduction of the lesion size in mice after experimental stroke (25). The precise role of autophagy as the third main mechanism of programmed cell death after ischemic stroke is not fully understood and remains controversial (26). However, cumulative evidence from preclinical in vitro and in vivo studies suggests a neuroprotective effect of pharmacological autophagy inhibition (2729).

Acute poststroke inflammation and stabilization of the BBB

Acute neuroinflammation after ischemic stroke is a highly complex sequence of events. In the following, we focus on three main factors and their role as targets in neuroprotective strategies, i.e., the activation of resident microglia, the opening of the BBB, and immune cell infiltration. Although microglia have been a subject of research for quite some time, the precise role of microglia under ischemic conditions is not yet known. Microglia are involved in both promoting ischemic brain damage and facilitating poststroke recovery. The latter depends on the state of function as expressed by microglia polarization (30). Thus, pharmacological modulation of microglial polarization is a promising neuroprotective strategy. For example, fingolimod, currently used in the treatment of multiple sclerosis, has been shown to modulate microglial polarization, leading to acute neuroprotection and enhanced angiogenesis in rodent stroke models (31, 32).

Inhibiting stroke-induced BBB breakdown, which plays a detrimental role in neuroinflammation and in the development of neurological dysfunctions, was identified as a possible therapeutic strategy (33). The main strategy for pharmacological stabilization of the BBB is the inhibition of matrix metalloproteases (MMP) which are responsible for the degradation of BBB components (34). Several preclinical neuroprotective agents such as resistin, lithium, and apelin-13 have been shown to stabilize the BBB as part of their neuroprotective mechanisms (3537). BBB breakdown is one important factor for the infiltration of peripheral immune cells, including neutrophils, T cells and B cells, as well as neutrophil-derived reactive oxygen species, cytokines, and proteases; they promote early neuroinflammation. Thus, neutrophils are examined as treatment targets by blocking neutrophil adhesion to endothelial cells, thus inhibiting the infiltration into the brain parenchyma (38). However, despite promising preclinical results by using adhesion blockers such as enlimomab, an anti-intercellular adhesion molecule 1 (ICAM-1) antibody, the clinical translation has failed (39). T cells were shown to have dual roles in stroke pathophysiology depending on their subtype. While CD4+, CD8+, and γδT cells seem to promote brain injury, Treg cells (regulatory T cells) have been shown to have neuroprotective effects (40, 41). Consistently, amplification of Treg cells by CD28SA leads to a reduction in infarct size in mouse model of stroke (42). In analogy to regulating T cells, Breg (regulatory B cells) are also involved in the poststroke inflammatory response (43). Again, the increase of Breg cells, with special emphasis on interleukin-10 producing subtypes, revealed protective effects in preclinical stroke models (44).

Transplantation of stem cells and delivery of extracellular vesicles (EVs)

Beside neuroprotective drugs, stem cell transplantation and stem cell-derived EVs have gained increasing interest in recent years. Several types of stem cells such as neural progenitor cells (NPC) and mesenchymal stem cells (MSC) have been tested in animal models of stroke, achieving promising beneficial effects (45). Stem cell transplantation has been proven safe for patients during the acute phase of stroke (46). Similar to pharmacological intervention targets, stem cell transplantation also affects several signaling pathways, such as BBB disruption and immune system response (47, 48). For instance, transplanting NPC into mice exposed to middle cerebral artery occlusion (MCAO) within 24 hours reduces infarct size and decreases BBB damage (49). After NPC transplantation, Evans blue dye and IgG leakage into the brain parenchyma are significantly reduced, suggesting BBB-stabilizing effects of NPC (49, 50). The BBB-stabilizing results from NPC are mainly achieved by inhibiting MMP activation and regulation of microglia activation (49, 50). Transplanted NPC can also affect peripheral immune organs, leading to a decrease of inflammatory cytokines such as TNF-α, IL-1β, IL-6, and Iba-1 (51). These beneficial effects are not limited to NPCs. MSCs, for instance, can also achieve the same effects (51). In contrast to common initial belief, however, the majority of transplanted stem cells are trapped in peripheral organs, suggesting that grafted stem cells stabilize the BBB and promote neural regeneration by indirect means (52). Such indirect means include paracrine factors. The latter became obvious when conditioned medium from stem cells was found to be as therapeutically effective as stem cells themselves (5355).

Whereas initial work claimed soluble factors to be the biological mediators of the aforementioned effects, recent work has resulted in the concept of EVs (56, 57). The latter are a heterogeneous group of vesicles in the nanometer range that include exosomes and microvesicles, to name a few (58). In this context, EVs are capable of transporting a wide variety of molecules and mediators like proteins (e.g., cell adhesion molecules, signal molecules, membrane organizing proteins), nucleic acids (e.g., microRNAs-[miRs]), as well as lipids (59). EVs are critically involved in intercellular communication under both physiological and pathophysiological conditions, supporting cell protection, cell regeneration, as well as immune modulation (57, 59). The application of stem cell-derived EVs therefore offers a great opportunity for stroke treatment. Moreover, EV administration appears to be safe in mammals, including humans, thus avoiding potential side effects of stem cell transplantation, in particular the risk of malignant stem cell transformation (57, 60).

Preclinical research has shown that the therapeutic potential of EVs is not inferior to stem cell transplantation (6164). Compared to stem cells, stem cell-derived EVs are easier to obtain and face fewer ethical issues. Such EVs affect multiple stroke-associated aspects, including angiogenesis, neurogenesis, and neuroprotection (65). Recent evidence suggests that miRs inside EVs are key players of EV-induced neuroprotection (66). Among these, miR-21a, miR-26a, and miR-126 are associated with strong and robust effects in preclinical stroke models, as they explicitly reduce infarct volume and stimulate neurogenesis as well as angiogenesis (6769).

Nevertheless, both stem cell transplantation and EV delivery under stroke conditions predominantly focus on subacute or even chronic stages of the disease. Hence, only a few studies analyzed the impact of stem cells during the acute phase of stroke. A phase II clinical trial (MASTERS, NCT01436487) by Hess and colleagues on patients with acute ischemic stroke, however, revealed no significant difference in global stroke recovery after intravenous treatment with multipotent progenitor cells between 24 and 48 hours after ischemic stroke (46). This is followed by a phase III clinical trial (MASTERS-2, NCT03545607) which investigates treatment with adult stem cells given intravenously 18-36 hours after stroke (MASTERS-2, NCT03545607). For the latter, results are not available yet. The therapeutic potential of stem cells and EVs alike are further discussed in the paragraph on chronic stroke.

SUBACUTE PHASE OF STROKE

In the acute phase of stroke, cell death, inflammation and scarring processes start within minutes to hours after stroke onset. The subacute phase is characterized by tissue reorganization. However, this process is limited by decreasing endogenous plasticity of the lesioned brain tissue. Whereas stroke leads to oxidative stress, excitotoxicity, reperfusion injury, and inflammation, poststroke recovery requires the stimulation of neuronal circuits and the induction of molecular growth programs in the brain (70). Interestingly, axonal sprouting, neurogenesis, gliogenesis and angiogenesis share common features like structural growth as well as interactions with other cells. However, these molecular mechanisms only represent transient regenerative cellular niches for neural repair after stroke (70). Li and colleagues report a significantly reduced gene regulation of the axonal sprouting connectome three weeks after stroke, indicating the loss of a coordinated growth state and a molecular closure of the sensitive period in the subacute phase after stroke (71). The global goal of stroke treatment therefore is to increase either the amount or the duration of endogenous plasticity.

Immunological aspects

The acute effects of ischemic stroke also lead to the activation of microglia and astrocytes which secrete proinflammatory factors, further recruiting peripheral immune cells to the ischemic area (72, 73). Additionally, dying neurons themselves, astrocytes, and endothelial cells release danger-associated molecular patterns (DAMPs), which activate microglia as well as peripheral immune cells in a detrimental cascade (72, 73).

Under physiological conditions, microglia are the innate immune cells of the brain, in their resting state constantly surveilling their environment and contributing to tissue homeostasis (74). Ischemic injury initiates the modulation of the resting microglia to an activated state as an immediate response. In the acute phase after stroke, activated microglia mainly represent the M1 type, inducing proinflammatory effects with detrimental consequences for the neurons in the ischemic penumbra (75, 76). In the subacute phase of stroke, however, the microglia population further shifts to the M2 type with an antiinflammatory purpose (77). This M1/M2 shift is often referred to as a double-edged sword action of microglia during stroke, which can be both beneficial and detrimental. Recent transcriptomic analyses of activated microglia display an activation of pathways outside the classic M1/M2 polarization paradigm, making further invesigations regarding this heterogeneity recommendable (74).

After the initial accumulation of microglia, other immune cells such as macrophages, lymphocytes, dendritic cells, and neutrophils infiltrate the lesion site (78). The latter are the main cause for the BBB breakdown after stroke (38). Whereas some studies claim that microglia overall seem to have largely protective effects after stroke, T lymphocytes of the innate immune system are regarded to be highly detrimental (79). On the other hand, Tregs of the adaptive immune system are thought to have protective effects. Tregs invade the ischemic tissue after the acute phase of stroke and act as neuroprotectors via the secretion of IL-10 (78). However, the exact mode of function of Tregs after stroke needs to be further elucidated.

B lymphocytes have also been described to have both detrimental and protective properties after stroke (79). Doyle and collegues report that B cells can be detected in the affected brain tissue in mouse models of stroke as well as in human patients (80). The authors link this finding to the later development of dementia after stroke (80). Protective effects are rather mediated by Bregs and, in accordance to T-cells, the secretion of IL-10 plays a major role against inflammation and resulting neurologic deficits (8183). IL-10 inhibits a variety of T-cell proinflammatory responses and increases the population of Tregs. Furthermore, latest research discovered additional molecules secreted by Bregs to regulate immune responses, like TGF-β (transforming growth factor beta), IL-35 and granzyme B, as well as surface proteins like CD1d and PD-L1 (84). However, more studies are needed to unravel these novel pathways, as the understanding of Bregs function after stroke appears to be a promising treatment strategy for transfer into the clinics. However, the dual role of inflammation after ischemic stroke, affecting both injury and repair, remains challenging, and therapeutic approaches targeting several cell types at different time points after stroke appear most promising (72). An overview of the different immune cells being involved in the pathology of stroke is given in Figure 2.

Fig 2

Figure 2. An overview of the different immune cells involved in the pathology of stroke. Neurons, astrocytes and endothelial cells release danger-associated molecular patterns (DAMPs) after stroke. DAMPs activate microglia and peripheral immune cells such as T cells and B cells, yielding a proinflammatory cytokine secretion profile from these cells. Meanwhile M2 microglia, regulatory T cells (Treg) and regulatory B cells (Breg) release antiinflammatory mediators that help reduce neuroinflammation. However, the dual role of inflammation after ischemic stroke is still unclear.

Gut-brain-axis

One of the largest compartments for immune cells in the body is the gastrointestinal tract (79). Here, commensal gut bacteria have direct contact to the intestinal epithelium as well as intestinal immune cells, supporting the host immune system with regard to peripheral immune education and homeostasis (79, 85, 86). The vital importance of these interactions, providing an intact immune system, was described more than half a century ago (87). More recently, the interactions between the gut microbiome and the immune system have been investigated with respect to stroke. Rosser and co-workers report that a disturbed gut microbiome negatively affects Bregs differentiation (88). Benakis and colleagues show that a disturbation of the gut microbiome by antibiotic treatment in the MCAO mouse model leads to an increase of Tregs and a simultaneous reduction of γδT-cells, correlating to decreased infarct size and increased behavioral outcome (89). However, a complete deletion of the gut microbiome had deleterious effects with regard to survival in the same model (90), pointing towards a sophisticated balance of the gut microbiome to provide beneficial effects. Furthermore, stroke itself has been shown to affect the gut microbiome composition via increased sympathetic activity, decreasing the intestinal motility, inducing dysbiosys and by that reducing microbial diversity (91). Taken together, the aforementioned studies indicate the existence of a bi-directional gut-brain-axis––an interaction of the gut microbiome via the intestinal immune system with the ischemic brain (Figure 3). After brain injury, the gut microbiome is altered and in turn modulates stroke outcome via modulation of postischemic inflammatory responses (92). Currently, the first clinical trials are conducted to further investigate how dysbiosis of the gut microbiome may influence immune response and outcome after stroke (93).

Fig 3

Figure 3. An overview of the gut-brain-axis after stroke. After brain injury, the gut microbiome is altered and in turn modulates stroke outcome via modulation of postischemic inflammatory responses. Stroke has been shown to affect the gut microbiome composition and diversity via increased sympathetic activity, decreasing the intestinal motility, and inducing dysbiosis. Selective removal of a certain population of intestinal bacteria has a beneficial effect on poststroke recovery. However, the complete removal of the intestinal flora has had a detrimental effect on stroke prognosis. The precise mechanisms of the gut-brain-axis after stroke are still unclear.

Neuroregenerative approaches using stem cells and EVs

The use of stem cells as a convalescent therapy after stroke has been established in animal models and is now transferred as a promising tool in human trials (94, 95). A 2019 Cochrane database analysis of randomized clinical trials of stem cell transplantation for ischemic stroke suggested an improvement in clinical outcome in patients (96). Nevertheless, the authors of the study concluded that the evidence base for an accurate assessment of stem cell therapy for the treatment of ischemic stroke is still insufficient, and further research in this field is thus urgently needed. Such research activities on both the neuroregenerative and the neuroprotective impact of stem cells in preclinical stroke models revealed paracrine mechanisms rather than cell transplantation to be the biological mediator in this respect (57). As stated before, these paracrine mechanisms gave rise to the concept of EVs. The current state of research with regard to molecular mediators transported by EVs is described in more detail below.

CHRONIC PHASE OF STROKE

The chronic phase of stroke displays distinct events such as the secondary loss of both glial cells and neurons as well as glial scar formation (97). The latter not only limits the stroke area but also blocks the synaptic connections between neurons (49). Along with this, an abnormal extracellular matrix (ECM) is formed, creating an unsuitable microenvironment for neurogenesis (98). Activation of microglia and astrocytes, as stated before, yields the production of proinflammatory cytokines that causes neuroinflammation and further deterioration of the microenvironment (99). Although BBB breakdown is a consequence of acute stroke in the first place, increased vessel leakage and risk of secondary hemorrhage can occur as a consequence of brain vasculature remodeling during the chronic stage of the disease (100). The aforementioned aspects limit neurological recovery during the chronic phase of stroke significantly. The current therapeutic approaches such as physical therapy programs and transcranial magnetic stimulation show beneficial effects for patients at the chronic phase of stroke (101, 102). However, the therapeutic effects of physiotherapy and magnetic stimulation are limited, depending on the severity of stroke lesions and the extent of neurological impairment (103). In this context, the persistence of endogenous neurogenesis within the adult mammalian brain offers new therapeutic targets during chronic stroke stages (92), albeit the majority of newborn cells would die within weeks (104). Stimulating poststroke endogenous neurogenesis by transplantion of exogenous stem cells may overcome these limitations (Figure 4).

Fig 4

Figure 4. Transplanting exogenous stem cells supports a beneficial stroke outcome. Several pathological processes are involved in the chronic stroke phase such as blood-brain barrier (BBB) breakdown, abnormal extracellular matrix formation, and proinflammatory factors secretion from microglia, creating a hostile microenvironment that hampers neurogenesis. Exogenous stem cell transplantation can inhibit microglia activation, increase the integrity of the BBB, and form a suitable microenvironment for neurogenesis. Interestingly, the majority of exogenous stem cells do not replace damaged tissue, suggesting that exogenous stem cells act in a paracrine way. MSCs, mesenchymal stem cells; NPCs, neural progenitor cells.

Transplantation of stem cells

Stereotactic administration of stem cells in the proximity of the stroke area is one possible option for delivering stem cells (105). The implanted stem cells can trigger the regenerative response within the ischemic tissue, by secreting various neurotrophic factors such as ECM-modifying enzymes (106). The latter contribute to remodeling the ECM in order to improve the microenvironment for synaptic regeneration. As described above, the formation of ECM limits the stroke volume but blocks the synaptic regeneration between newborn neurons. Intracerebral implantation of MSCs is considered to promote neurogenesis via remodeling the glial scar in this respect (107).

The activation of microglia and astrocytes that secrete proinflammatory cytokines such as TNF-α or IL-6 causes leukocyte mobilization in the blood and leukocyte infiltration in the brain tissue (108). These factors induce an inflammatory environment that is not conducive to neurogenesis. The reduction of inflammation levels in brain tissue due to cell therapies may also contribute to neurological recovery. Stem cells can regulate the immune response by producing antiinflammatory cytokines and thus decrease levels of activated microglia and macrophages, resulting in reduced secretion of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) (109). MSCs have been shown to reduce T-cell proliferation and activation, yielding decreased levels of proinflammatory cytokine production by monocytes/macrophages through cell-cell contact with immune cells (110, 111). Likewise, MSCs decrease the secretion of paracrine factors from microglia (such as TGF-β, indolamin-2,3-dioxygenase [IDO], and prostaglandin E2 [PGE2]), which results in the transition from a proinflammatory into an antiinflammatory state (112, 113). However, cell implantation into the periinfarct area is not a suitable method for clinical application. Other delivery methods such as systemic administration suffer from stem cell loss in peripheral organs (114, 115). However, these trapped stem cells are still beneficial in terms of enhancing poststroke neurological recovery and stimulating tissue regeneration, again demonstrating the role of paracrine mechanisms in stem cell therapy (45, 61).

Poststroke delivery of EVs

Unlike stem cells, the diameter of EVs is around 50 to 1000 nm, making EVs small enough to cross the BBB and directly affect the stroke area (56, 116). The mechanism of action of stem cell-derived EVs is mainly derived from the contents of EVs (Figure 5). The miR contents in stem cell-derived EVs have been proven to be neuroprotective or to induce neurogenesis at chronic stages of the disease (117). MSC-EV-derived miR clusters such as miR-17-92 have been proven to promote both angiogenesis and neurogenesis (118). More and more angiogenesis and neurogenesis-related miRs such as miR-133b, miR-134, and miR-181b are found in MSC-EVs in recent years (119), suggesting that miRs within EVs are involved in EV-induced angiogenesis and neurogenesis. Beside the miR content, proteins and cytokines are also involved in EV-induced neurogenesis and angiogenesis (117, 120, 121). Fibroblast growth factor 2 (FGF2), placental growth factor (PGF), and hepatocyte growth factor (HGF) that are important in adult neurogenesis are found in MSC-EVs (122). Cytokines that are related to angiogenesis, such as vascular endothelial growth factor-A (VEGF-A) and VEGF-C, are enriched in MSC-EVs as well (123). Such selected cytokines and miRs in stem cell-derived EVs support the EV-induced neurogenesis and angiogenesis after stroke (124). Beside stimulating neurogenesis and angiogenesis, EVs can also remodel the ECM (125). Evidence indicates that EVs contain ECM-related enzymes suggesting that EVs may be involved in ECM remodeling after stroke (125). MMP-9, as a key protein involved in ECM remodeling, has indeed been proven to be regulated by stem cell-derived EVs. Both MMP-9 and FGF-2 are found in mesangioblast stem cell-derived EVs (126). MMP-9 degrades gelatin within the ECM, whereas FGF-2 helps form the ECM, thus demonstrating a complex role of EVs in tissue regeneration.

Fig 5

Figure 5. Stem cell-derived EVs stimulate poststroke neuroregeneration. Stem cell-derived extracellular vesicles (EVs) yield similar effects as exogenous stem cells under stroke conditions. Stem cell-derived EVs stabilize the extracellular microenvironment and enhance the integrity of the blood-brain barrier (BBB). They also reduce the proinflammatory factors secretion of microglia, contributing to a restoration of the former abnormal extracellular matrix. Abbreviations: NPCs, neural progenitor cells; MSCs, mesenchymal stem cells.

Like stem cell transplantation, stem cell-derived EVs can regulate the immune responses of the recipient (127). Stem cell-derived EVs affect the immune system after stroke mainly by regulating microglia activation in the central nervous system (CNS) and peripheral immune cells such as T cells and B cells (128). In the CNS, the microglia acts as a double-edged sword in stroke pathology. In the acute phase, activated microglia are mainly M1 type, inducing pro-inflammatiory effects (76). At this phase, M1 microglia recruit the peripheral leukocytes to the brain to clean the damaged tissues in the stroke area and cause the increase of pro-apoptotic cytokines such as TNF-α and IL-6, leading to neuron loss in the ischemic penumbra (75). However, M2 microglia are also activated in the acute phase, but their number is low. M2 microglia play an antiinflammatory role in the subacute and chronic phase of stroke (77). The M2 microglia are the majority of microglia in the CNS that release antiinflammatory cytokines such as IL-10, which redcues the overall state of inflammation in the brain but may cause immunosuppression. The latter is a common phenomenon in chronic phase stroke patients, making them vulnerable to infectious diseases. The balance between M1 and M2 microglia is one of the key points of stroke recovery (74). Stem cell-derived EVs show significant effects on regulating microglia activation (129). Several antiinflammatory miRs and proteins have been identified in human iPSC-derived neural stem cell EVs (121). Eight antiinflammatory and neurogenesis-related miRs (miRs-320a, 320b, 103a-3p, 21–5p, 26a-5p, 30a-3p, 181a-5p, 191–5p) were identified in iPSC-EVs. Among them, miR-21 has been shown to have neuroprotective and antiinflammatory properties in traumatic brain injury and stroke (130). miR-21 mediates antiinflammatory activity through the downregulation of NF-κB and TNF-α and induction of the antiinflammatory cytokine IL-10 (131). Another miR, miR-126 from ADMSC-EVs, can promote functional recovery after stroke in rats by improving neurogenesis and suppressing microglia activation (27). Meanwhile, Gal-3BP, a suppressor of inflammatory responses through NF-κB pathway, was found in iPSC-EVs. The delivery of Gal-3BP into microglia was able to restrain neuroinflammation in Alzheimer’s disease (132).

The remodeling of brain vasculature with lower BBB integrity causes inefficient blood supply and increases the potential bleeding risk (100). Enhancement of neovascularization and strengthening of vascular links are therapeutic goals. Stem cell-derived EVs have shown the potential to increase BBB integrity and promote angiogenesis (41). Hence, MSC-EVs promote both angiogenesis and integrity of the BBB under chronic stroke conditions. These effects are, however, not exclusive to MSC-EVs alone. As a matter of fact, NPC-EVs also stabilize BBB integrity by decreasing NF-κB activation levels via altered ABCB1 expression patterns on the luminal endothelium (128). Along with this, EVs from endothelial cells decrease PTEN expression, stimulate AKT phosphorylation, and increase tight junction protein expression in cells, which may also contribute to enhanced BBB stability (133). Although EVs are able to enhance the integrity of the BBB under preclinical stroke settings (134), the precise mechanisms remain unclear. Further studies are therefore needed in this respect.

Stem cells and EVs under clinical stroke settings

The results of preclinical studies have encouraged the extension of stem cell transplantation and EVs to the clinic (135). Several early phase I trials have shown the safety of MSC transplantation (136). A long-term follow-up study of autologous MSC transplantation in stroke patients shows improved recovery after stroke (137). The study included 85 patients with acute (<72 hours after onset) nonlacunar infarction within the middle cerebral artery territory who were followed up for six months. The patients were systemically administered with 5 × 107 autologous MSCs at 5 weeks and 7 weeks after stroke. SDF-1α and CXCL 12, proteins associated with MSC homing, were increased in the serum after MSC transplantation. The modified Rankin Scale (mRS) score was decreased in the MSC transplantation patients. The study proved the safety and beneficial effect of MSC transplantation. Further research showed that different dosages of MSC transplantation are safe and efficient (138). In the phase I stage, three doses (0.5, 1.0, and 1.5 million cells/kg body weight) were tested (n = 5 per cohort), and all dosages were found to be safe (138). In the phase II stage, 1.5 million cells/kg were intravenously administered to 21 patients (138). Significant functional outcome improvement was observed in 35.5% of patients at 6 months and 12 months post-transplantation. However, there are some clinical trials indicating that MSC transplantation may not affect clinical outcomes at all. A Phase I/II trial of intravenously administered autologous MSCs to 20 patients in the chronic phase of stroke between 3 months and 2 years following stroke onset did not show significant differences between the transplantation and control groups in terms of functional outcome (139). In conclusion, the completed or ongoing MSC transplantation clinical trials proved the safety of stem cell transplantation, but the effectiveness of MSC transplantation is still under debate. The data on EVs under clinical stroke settings is scarce. Only one phase I/II clinical trial currently investigates the role of allogenic MSC-derived EVs transfected with miR-124 in stroke patients (NCT03384433). The study is still ongoing, and no results are available yet.

CONCLUSION

Although recent progress has been made in modern stroke treatment, many stroke patients do not gain any benefit from it because of therapeutic limitations. Adjuvant novel treatment paradigms are therefore in order. Such experimental strategies focusing on acute therapeutic interventions primarily include neuroprotection, which has failed in clinical trials until recently. The scientific interest has now shifted towards subacute and chronic stroke stages. Transplantations of stem cells have been thoroughly investigated under such stroke conditions. However, stem cells are not integrated into residing neural networks but act via EV secretion. EV-based strategies may therefore present a next-generation therapeutic tool in stroke treatment. The advantage of such systems include the ability of EVs to cross both cell membranes and the BBB under physiological and pathological conditions alike. To the best of current knowledge, EVs have little toxic effects and no risk of tumor formation (140, 141). Serving as nanoparticle carrier, they can induce pleiotropic effects in their target organs. Nevertheless, additional studies are necessary for addressing fundamental issues with regard to technical and biological features.

Conflict of Interest: The authors declare no potential conflict of interest with respect to research, authorship and/or publication of this chapter.

Copyright and Permission Statement: The authors confirm that the materials included in this chapter do not violate copyright laws. Where relevant, appropriate permissions have been obtained from the original copyright holder(s), and all original sources have been appropriately acknowledged or referenced.

REFERENCES

  1. Wu P, Zeng F, Li YX, Yu BL, Qiu LH, Qin W, et al. Changes of resting cerebral activities in subacute ischemic stroke patients. Neural Regen Res 2015;10:760–5. https://doi.org/10.4103/1673-5374.156977
  2. Kamalian S, Lev MH. Stroke Imaging. Radiol Clin North Am 2019;57:717–32. https://doi.org/10.1016/j.rcl.2019.02.001
  3. Kamtchum-Tatuene J, Jickling GC. Blood Biomarkers for Stroke Diagnosis and Management. NeuroMolecular Med 2019;21:344–68. https://doi.org/10.1007/s12017-019-08530-0
  4. Makris K, Haliassos A, Chondrogianni M, Tsivgoulis G. Blood biomarkers in ischemic stroke: potential role and challenges in clinical practice and research. Crit Rev Clin Lab Sci 2018;55:294–328. https://doi.org/10.1080/10408363.2018.1461190
  5. Campbell BCV, Khatri P. Stroke. Lancet 2020;396:129–42. https://doi.org/10.1016/S0140-6736(20)31179-X
  6. O’Collins VE, Macleod MR, Donnan GA, Horky LL, Van Der Worp BH, Howells DW. 1,026 Experimental treatments in acute stroke. Ann Neurol 2006;59:467–77. https://doi.org/10.1002/ana.20741
  7. Dirnagl U, Iadecola C, Moskowitz M a. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci 1999;22:391–7. https://doi.org/10.1016/S0166-2236(99)01401-0
  8. Patel RAG, McMullen PW. Neuroprotection in the treatment of acute ischemic stroke. Prog Cardiovasc Dis 2017;59:542–8. https://doi.org/10.1016/j.pcad.2017.04.005
  9. Lai TW, Shyu WC, Wang YT. Stroke intervention pathways: NMDA receptors and beyond. Trends Mol Med 2011;17:266–75. https://doi.org/10.1016/j.molmed.2010.12.008
  10. Lai TW, Zhang S, Wang YT. Excitotoxicity and stroke: Identifying novel targets for neuroprotection. Prog Neurobiol 2014;115:157–88. https://doi.org/10.1016/j.pneurobio.2013.11.006
  11. Wu QJ, Tymianski M. Targeting nmda receptors in stroke: New hope in neuroprotection Tim Bliss. Mol Brain 2018;11. https://doi.org/10.1186/s13041-018-0357-8
  12. Allen CL, Bayraktutan U. Oxidative stress and its role in the pathogenesis of ischaemic stroke. Int J Stroke 2009;4:461–70. https://doi.org/10.1111/j.1747-4949.2009.00387.x
  13. Fabian RH, DeWitt DS, Kent TA. In vivo detection of superoxide anion production by the brain using a cytochrome c electrode. J Cereb Blood Flow Metab 1995;15:242–7. https://doi.org/10.1038/jcbfm.1995.30
  14. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 2007;87:315–424. https://doi.org/10.1152/physrev.00029.2006
  15. Sun MS, Jin H, Sun X, Huang S, Zhang FL, Guo ZN, et al. Free Radical Damage in Ischemia-Reperfusion Injury: An Obstacle in Acute Ischemic Stroke after Revascularization Therapy. Oxid Med Cell Longev 2018;2018. https://doi.org/10.1155/2018/3804979
  16. Son JW, Choi H, Yoo A, Park HH, Kim YS, Lee KY, et al. Activation of the phosphatidylinositol 3-kinase pathway plays important roles in reduction of cerebral infarction by cilnidipine. J Neurochem 2015;135:186–93. https://doi.org/10.1111/jnc.13254
  17. Gupta S, Sharma U, Jagannathan NR, Gupta YK. Neuroprotective effect of lercanidipine in middle cerebral artery occlusion model of stroke in rats. Exp Neurol 2017;288:25–37. https://doi.org/10.1016/j.expneurol.2016.10.014
  18. Maniskas ME, Roberts JM, Aron I, Fraser JF, Bix GJ. Stroke neuroprotection revisited: Intra-arterial verapamil is profoundly neuroprotective in experimental acute ischemic stroke. J Cereb Blood Flow Metab 2015;36:721–30. https://doi.org/10.1177/0271678X15608395
  19. Zhang Y, Qiao L, Xu W, Wang X, Li H, Xu W, et al. Paeoniflorin attenuates cerebral ischemia-induced injury by regulating Ca2+/CaMKII/CREB signaling pathway. Molecules 2017;22. https://doi.org/10.3390/molecules22030359
  20. Radak D, Katsiki N, Resanovic I, Jovanovic A, Sudar-Milovanovic E, Zafirovic S, et al. Apoptosis and Acute Brain Ischemia in Ischemic Stroke. Curr Vasc Pharmacol 2016;15:115–22. https://doi.org/10.2174/1570161115666161104095522
  21. Sekerdag E, Solaroglu I, Gursoy-Ozdemir Y. Cell Death Mechanisms in Stroke and Novel Molecular and Cellular Treatment Options. Curr Neuropharmacol 2018;16:1396–415. https://doi.org/10.2174/1570159X16666180302115544
  22. Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduct 2015;35:600–4. https://doi.org/10.3109/10799893.2015.1030412
  23. Karatas H, Aktas Y, Gursoy-Ozdemir Y, Bodur E, Yemisci M, Caban S, et al. A nanomedicine transports a peptide caspase-3 inhibitor across the blood-brain barrier and provides neuroprotection. J Neurosci 2009;29:13761–9. https://doi.org/10.1523/JNEUROSCI.4246-09.2009
  24. Jun-Long H, Yi L, Bao-Lian Z, Jia-Si L, Ning Z, Zhou-Heng Y, et al. Necroptosis Signaling Pathways in Stroke: From Mechanisms to Therapies. Curr Neuropharmacol 2018;16:1327–39. https://doi.org/10.2174/1570159X16666180416152243
  25. Cruz SA, Qin Z, Stewart AFR, Chen HH. Dabrafenib, an inhibitor of RIP3 kinase-dependent necroptosis, reduces ischemic brain injury. Neural Regen Res 2018;13:252–6. https://doi.org/10.4103/1673-5374.226394
  26. Nabavi SF, Sureda A, Sanches-Silva A, Pandima Devi K, Ahmed T, Shahid M, et al. Novel therapeutic strategies for stroke: The role of autophagy. Crit Rev Clin Lab Sci 2019;56:182–99. https://doi.org/10.1080/10408363.2019.1575333
  27. Tang T, Gao D, Yang X, Hua X, Li S, Sun H. Exogenous Netrin-1 Inhibits Autophagy of Ischemic Brain Tissues and Hypoxic Neurons via PI3K/mTOR Pathway in Ischemic Stroke. J Stroke Cerebrovasc Dis 2019;28:1338–45. https://doi.org/10.1016/j.jstrokecerebrovasdis.2019.01.032
  28. Baek SH, Noh AR, Kim KA, Akram M, Shin YJ, Kim ES, et al. Modulation of mitochondrial function and Autophagy mediates Carnosine Neuroprotection against ischemic brain damage. Stroke 2014;45:2438–43. https://doi.org/10.1161/STROKEAHA.114.005183
  29. Zhou H, Wang J, Jiang J, Stavrovskaya IG, Li M, Li W, et al. N-acetyl-serotonin offers neuroprotection through inhibiting mitochondrial death pathways and autophagic activation in experimental models of ischemic injury. J Neurosci 2014;34:2967–78. https://doi.org/10.1523/JNEUROSCI.1948-13.2014
  30. Jiang CT, Wu WF, Deng YH, Ge JW. Modulators of microglia activation and polarization in ischemic stroke (Review). Mol Med Rep 2020;21:2006–18. https://doi.org/10.3892/mmr.2020.11003
  31. Shang K, He J, Zou J, Qin C, Lin L, Zhou LQ, et al. Fingolimod promotes angiogenesis and attenuates ischemic brain damage via modulating microglial polarization. Brain Res 2020;1726. https://doi.org/10.1016/j.brainres.2019.146509
  32. Qin C, Fan WH, Liu Q, Shang K, Murugan M, Wu LJ, et al. Fingolimod protects against ischemic white matter damage by modulating microglia toward M2 polarization via STAT3 pathway. Stroke 2017;48:3336–46. https://doi.org/10.1161/STROKEAHA.117.018505
  33. Sifat AE, Vaidya B, Abbruscato TJ. Blood-Brain Barrier Protection as a Therapeutic Strategy for Acute Ischemic Stroke. AAPS J 2017;19:957–72. https://doi.org/10.1208/s12248-017-0091-7
  34. Kurzepa J, Kurzepa J, Golab P, Czerska S, Bielewicz J. The significance of matrix metalloproteinase (MMP)-2 and MMP-9 in the ischemic stroke. Int J Neurosci 2014;124:707–16. https://doi.org/10.3109/00207454.2013.872102
  35. Haupt M, Zechmeister B, Bosche B, Lieschke S, Zheng X, Zhang L, et al. Lithium enhances post-stroke blood-brain barrier integrity, activates the MAPK/ERK1/2 pathway and alters immune cell migration in mice. Neuropharmacology 2020;181:108357. https://doi.org/10.1016/j.neuropharm.2020.108357
  36. Chu H, Yang X, Huang C, Gao Z, Tang Y, Dong Q. Apelin-13 Protects against Ischemic Blood-Brain Barrier Damage through the Effects of Aquaporin-4. Cerebrovasc Dis 2017;44:10–25. https://doi.org/10.1159/000460261
  37. Behrouzifar S, Vakili A, Bandegi AR, Kokhaei P. Neuroprotective nature of adipokine resistin in the early stages of focal cerebral ischemia in a stroke mouse model. Neurochem Int 2018;114:99–107. https://doi.org/10.1016/j.neuint.2018.02.001
  38. Jickling GC, Liu DZ, Ander BP, Stamova B, Zhan X, Sharp FR. Targeting neutrophils in ischemic stroke: Translational insights from experimental studies. J Cereb Blood Flow Metab 2015;35:888–901. https://doi.org/10.1038/jcbfm.2015.45
  39. Sherman D, Bes A, Easton JD, Hacke W, Kaste M, Polmar SH, et al. Use of anti-ICAM-1 therapy in ischemic stroke: Results of the enlimomab acute stroke trial. Neurology 2001;57:1428–34. https://doi.org/10.1212/WNL.57.8.1428
  40. Kim JY, Park J, Chang JY, Kim S-H, Lee JE. Inflammation after Ischemic Stroke: The Role of Leukocytes and Glial Cells. Exp Neurobiol 2016;25:241–51. https://doi.org/10.5607/en.2016.25.5.241
  41. Wang J, Chen S, Zhang W, Chen Y, Bihl JC. Exosomes from miRNA-126-modified endothelial progenitor cells alleviate brain injury and promote functional recovery after stroke. CNS Neurosci Ther 2020;26:1255–65. https://doi.org/10.1111/cns.13455
  42. Na SY, Mracsko E, Liesz A, Hünig T, Veltkamp R. Amplification of regulatory T cells using a CD28 superagonist reduces brain damage after ischemic stroke in mice. Stroke 2015;46:212–20. https://doi.org/10.1161/STROKEAHA.114.007756
  43. Lebien TW, Tedder TF. B lymphocytes: How they develop and function. Blood 2008;112:1570–80. https://doi.org/10.1182/blood-2008-02-078071
  44. Seifert HA, Vandenbark AA, Offner H. Regulatory B cells in experimental stroke. Immunology 2018;154:169–77. https://doi.org/10.1111/imm.12887
  45. Chrostek MR, Fellows EG, Crane AT, Grande AW, Low WC. Efficacy of stem cell-based therapies for stroke. Brain Res 2019;1722. https://doi.org/10.1016/j.brainres.2019.146362
  46. Hess DC, Wechsler LR, Clark WM, Savitz SI, Ford GA, Chiu D, et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol 2017;16:360–8. https://doi.org/10.1016/S1474-4422(17)30046-7
  47. Boese AC, Le QSE, Pham D, Hamblin MH, Lee JP. Neural stem cell therapy for subacute and chronic ischemic stroke. Stem Cell Res Ther 2018;9. https://doi.org/10.1186/s13287-018-0913-2
  48. Saft M, Gonzales-Portillo B, Park YJ, Cozene B, Sadanandan N, Cho J, et al. Stem Cell Repair of the Microvascular Damage in Stroke. Cells 2020;9. https://doi.org/10.3390/cells9092075
  49. Huang L, Wu ZB, ZhuGe Q, Zheng WM, Shao B, Wang B, et al. Glial scar formation occurs in the human brain after ischemic stroke. Int J Med Sci 2014;11:344–8. https://doi.org/10.7150/ijms.8140
  50. Eckert A, Huang L, Gonzalez R, Kim H-S, Hamblin MH, Lee J-P. Bystander Effect Fuels Human Induced Pluripotent Stem Cell-Derived Neural Stem Cells to Quickly Attenuate Early Stage Neurological Deficits After Stroke. Stem Cells Transl Med 2015;4:841–51. https://doi.org/10.5966/sctm.2014-0184
  51. Trounson A, McDonald C. Stem Cell Therapies in Clinical Trials: Progress and Challenges. Cell Stem Cell 2015;17:11–22. https://doi.org/10.1016/j.stem.2015.06.007
  52. Zhou G, Wang Y, Gao S, Fu X, Cao Y, Peng Y, et al. Potential Mechanisms and Perspectives in Ischemic Stroke Treatment Using Stem Cell Therapies. Front Cell Dev Biol 2021;9:646927. https://doi.org/10.3389/fcell.2021.646927
  53. Doeppner TR, Traut V, Heidenreich A, Kaltwasser B, Bosche B, Bähr M, et al. Conditioned Medium Derived from Neural Progenitor Cells Induces Long-term Post-ischemic Neuroprotection, Sustained Neurological Recovery, Neurogenesis, and Angiogenesis. Mol Neurobiol 2017;54:1531–40. https://doi.org/10.1007/s12035-016-9748-y
  54. Faezi M, Nasseri Maleki S, Aboutaleb N, Nikougoftar M. The membrane mesenchymal stem cell derived conditioned medium exerts neuroprotection against focal cerebral ischemia by targeting apoptosis. J Chem Neuroanat 2018;94:21–31. https://doi.org/10.1016/j.jchemneu.2018.08.004
  55. Tsai MJ, Tsai SK, Hu BR, Liou DY, Huang SL, Huang MC, et al. Recovery of neurological function of ischemic stroke by application of conditioned medium of bone marrow mesenchymal stem cells derived from normal and cerebral ischemia rats. J Biomed Sci 2014;21. https://doi.org/10.1186/1423-0127-21-5
  56. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018;7. https://doi.org/10.1080/20013078.2018.1461450
  57. Willis CM, Nicaise AM, Peruzzotti-Jametti L, Pluchino S. The neural stem cell secretome and its role in brain repair. Brain Res 2020;1729. https://doi.org/10.1016/j.brainres.2019.146615
  58. Yáñez-Mó M, Siljander PRM, Andreu Z, Zavec AB, Borràs FE, Buzas EI, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles 2015;4:1–60. https://doi.org/10.3402/jev.v4.27066
  59. Doeppner TR, Bähr M, Giebel B, Hermann DM. Immunological and non-immunological effects of stem cell-derived extracellular vesicles on the ischaemic brain. Ther Adv Neurol Disord 2018;11. https://doi.org/10.1177/1756286418789326
  60. Doeppner TR, Bähr M, Hermann DM, Giebel B. Concise Review: Extracellular Vesicles Overcoming Limitations of Cell Therapies in Ischemic Stroke. Stem Cells Transl Med 2017;6:2044–52. https://doi.org/10.1002/sctm.17-0081
  61. Doeppner TR, Herz J, Görgens A, Schlechter J, Ludwig A-K, Radtke S, et al. Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression. Stem Cells Transl Med 2015;4:1131–43. https://doi.org/10.5966/sctm.2015-0078
  62. Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab 2013;33:1711–5. https://doi.org/10.1038/jcbfm.2013.152
  63. Xin H, Liu Z, Buller B, Li Y, Golembieski W, Gan X, et al. MiR-17–9. enriched exosomes derived from multipotent mesenchymal stromal cells enhance axon-myelin remodeling and motor electrophysiological recovery after stroke. J Cereb Blood Flow Metab 2021;41:1131–44. https://doi.org/10.1177/0271678X20950489
  64. Zheng X, Zhang L, Kuang Y, Venkataramani V, Jin F, Hein K, et al. Extracellular Vesicles Derived from Neural Progenitor Cells--a Preclinical Evaluation for Stroke Treatment in Mice. Transl Stroke Res 2021;12:185–203. https://doi.org/10.1007/s12975-020-00814-z
  65. Xia Y, Ling X, Hu G, Zhu Q, Zhang J, Li Q, 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. https://doi.org/10.1186/s13287-020-01834-0
  66. Terriaca S, Fiorelli E, Scioli MG, Fabbri G, Storti G, Cervelli V, et al. Endothelial progenitor cell-derived extracellular vesicles: Potential therapeutic application in tissue repair and regeneration. Int J Mol Sci 2021;22. https://doi.org/10.3390/ijms22126375
  67. Ma Y, Li C, Huang Y, Wang Y, Xia X, Zheng JC. Exosomes released from neural progenitor cells and induced neural progenitor cells regulate neurogenesis through miR-21a. Cell Commun Signal 2019;17. https://doi.org/10.1186/s12964-019-0418-3
  68. Chen H, He Y, Chen S, Qi S, Shen J. Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: Applications for natural product efficacy with omics and systemic biology. Pharmacol Res 2020;158. https://doi.org/10.1016/j.phrs.2020.104877
  69. Ling X, Zhang G, Xia Y, Zhu Q, Zhang J, Li Q, et al. Exosomes from human urine-derived stem cells enhanced neurogenesis via miR-26a/HDAC6 axis after ischaemic stroke. J Cell Mol Med 2020;24:640–54. https://doi.org/10.1111/jcmm.14774
  70. Brumm AJ, Carmichael ST. Not just a rush of blood to the head. Nat Med 2012;18:1609-10. https://doi.org/10.1038/nm.2990
  71. Li S, Overman JJ, Katsman D, Kozlov S V., Donnelly CJ, Twiss JL, et al. An age-related sprouting transcriptome provides molecular control of axonal sprouting after stroke. Nat Neurosci 2010;13:1496–506. https://doi.org/10.1038/nn.2674
  72. Lambertsen KL, Finsen B, Clausen BH. Post-stroke inflammation-target or tool for therapy? Acta Neuropathol 2019;137:693–714. https://doi.org/10.1007/s00401-018-1930-z
  73. Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: Friend and foe for ischemic stroke. J Neuroinflammation 2019;16. https://doi.org/10.1186/s12974-019-1516-2
  74. Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol 2016;173:649–65. https://doi.org/10.1111/bph.13139
  75. Anttila JE, Whitaker KW, Wires ES, Harvey BK, Airavaara M. Role of microglia in ischemic focal stroke and recovery: focus on Toll-like receptors. Prog Neuro-Psychopharmacology Biol Psychiatry 2017;79:3–14. https://doi.org/10.1016/j.pnpbp.2016.07.003
  76. Zhao SC, Ma LS, Chu ZH, Xu H, Wu WQ, Liu F. Regulation of microglial activation in stroke. Acta Pharmacol Sin 2017;38:445–58. https://doi.org/10.1038/aps.2016.162
  77. Lee Y, Lee SR, Choi SS, Yeo HG, Chang KT, Lee HJ. Therapeutically targeting neuroinflammation and microglia after acute ischemic stroke. Biomed Res Int 2014;2014. https://doi.org/10.1155/2014/297241
  78. Selvaraj UM, Stowe AM. Long-term T cell responses in the brain after an ischemic stroke. Discov Med 2017;24:323–33.
  79. Anrather J, Iadecola C. Inflammation and stroke: An overview. Neurotherapeutics 2016;13:661-70. https://doi.org/10.1007/s13311-016-0483-x
  80. Doyle KP, Quach LN, Solé M, Axtell RC, Nguyen TV V., Soler-Llavina GJ, et al. B-lymphocyte-mediated delayed cognitive impairment following stroke. J Neurosci 2015;35:2133–45. https://doi.org/10.1523/JNEUROSCI.4098-14.2015
  81. Ren X, Akiyoshi K, Dziennis S, Vandenbark AA, Herson PS, Hurn PD, et al. Regulatory B cells limit CNS inflammation and neurologic deficits in murine experimental stroke. J Neurosci 2011;31:8556–63. https://doi.org/10.1523/JNEUROSCI.1623-11.2011
  82. Offner H, Hurn PD. A Novel Hypothesis: Regulatory B Lymphocytes Shape Outcome from Experimental Stroke. Transl Stroke Res 2012;3:324–30. https://doi.org/10.1007/s12975-012-0187-4
  83. Bodhankar S, Chen Y, Vandenbark AA, Murphy SJ, Offner H. IL-10-producing B-cells limit CNS inflammation and infarct volume in experimental stroke. Metab Brain Dis 2013;28:375-86. https://doi.org/10.1007/s11011-013-9413-3
  84. Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, et al. Immunosuppressive Mechanisms of Regulatory B Cells. Front Immunol 2021;12. https://doi.org/10.3389/fimmu.2021.611795
  85. Nishio J, Honda K. Immunoregulation by the gut microbiota. Cell Mol Life Sci 2012;69:3635-50. https://doi.org/10.1007/s00018-012-0993-6
  86. Kuhn KA, Stappenbeck TS. Peripheral education of the immune system by the colonic microbiota. Semin Immunol 2013;25:364–9. https://doi.org/10.1016/j.smim.2013.10.002
  87. Bauer H, Horowitz RE, Levenson SM, Popper H. The response of the lymphatic tissue to the microbial flora. Studies on germfree mice. Am J Pathol 1963;42:471-83.
  88. Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA, et al. Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production. Nat Med 2014;20:1334–9. https://doi.org/10.1038/nm.3680
  89. Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med 2016;22:516–23. https://doi.org/10.1038/nm.4068
  90. Winek K, Engel O, Koduah P, Heimesaat MM, Fischer A, Bereswill S, et al. Depletion of Cultivatable Gut Microbiota by Broad-Spectrum Antibiotic Pretreatment Worsens Outcome After Murine Stroke. Stroke 2016;47:1354–63. https://doi.org/10.1161/STROKEAHA.115.011800
  91. Houlden A, Goldrick M, Brough D, Vizi ES, Lénárt N, Martinecz B, et al. Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production. Brain Behav Immun 2016;57:10–20. https://doi.org/10.1016/j.bbi.2016.04.003
  92. Rahman AA, Amruta N, Pinteaux E, Bix GJ. Neurogenesis After Stroke: A Therapeutic Perspective. Transl Stroke Res 2021;12. https://doi.org/10.1007/s12975-020-00841-w
  93. Liu F, Cheng X, Zhong S, Liu C, Jolkkonen J, Zhang X, et al. Communications Between Peripheral and the Brain-Resident Immune System in Neuronal Regeneration After Stroke. Front Immunol 2020;11. https://doi.org/10.3389/fimmu.2020.01931
  94. Griauzde J, Ravindra VM, Chaudhary N, Gemmete JJ, Pandey AS. Neuroprotection for ischemic stroke in the endovascular era: A brief report on the future of intra-arterial therapy. J Clin Neurosci 2019;69:289–91. https://doi.org/10.1016/j.jocn.2019.08.001
  95. Phipps MS, Cronin CA. Management of acute ischemic stroke. BMJ 2020;368. https://doi.org/10.1136/bmj.l6983
  96. Boncoraglio GB, Ranieri M, Bersano A, Parati EA, Del Giovane C. Stem cell transplantation for ischemic stroke. Cochrane Database Syst Rev 2019;2019. https://doi.org/10.1002/14651858.CD007231.pub3
  97. Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial Cells: Role of the Immune Response in Ischemic Stroke. Front Immunol 2020;11:294. https://doi.org/10.3389/fimmu.2020.00294
  98. Kazanis I, Ffrench-Constant C. Extracellular matrix and the neural stem cell niche. Dev Neurobiol 2011;71:1006–17. https://doi.org/10.1002/dneu.20970
  99. Tjalkens RB, Popichak KA, Kirkley KA. Inflammatory Activation of Microglia and Astrocytes in Manganese Neurotoxicity. Adv. Neurobiol., vol. 18, Springer New York LLC; 2017, p. 159–81. https://doi.org/10.1007/978-3-319-60189-2_8
  100. Yang Y, Torbey MT. Angiogenesis and Blood-Brain Barrier Permeability in Vascular Remodeling after Stroke. Curr Neuropharmacol 2020;18:1250–65. https://doi.org/10.2174/1570159X18666200720173316
  101. Dobkin BH, Dorsch A. New evidence for therapies in stroke rehabilitation topical collection on cardiovascular disease and stroke. Curr Atheroscler Rep 2013;15. https://doi.org/10.1007/s11883-013-0331-y
  102. Fisicaro F, Lanza G, Grasso AA, Pennisi G, Bella R, Paulus W, et al. Repetitive transcranial magnetic stimulation in stroke rehabilitation: review of the current evidence and pitfalls. Ther Adv Neurol Disord 2019;12. https://doi.org/10.1177/1756286419878317
  103. Sehatzadeh S. Effect of increased intensity of physiotherapy on patient outcomes after stroke: An evidence-based analysis. Ont Health Technol Assess Ser 2015;15.
  104. Lindvall O, Kokaia Z. Neurogenesis following stroke affecting the adult brain. Cold Spring Harb Perspect Biol 2015;7. https://doi.org/10.1101/cshperspect.a019034
  105. Singh M, Pandey PK, Bhasin A, Padma M V., Mohanty S. Application of Stem Cells in Stroke: A Multifactorial Approach. Front Neurosci 2020;14. https://doi.org/10.3389/fnins.2020.00473
  106. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2014;15:786–801. https://doi.org/10.1038/nrm3904
  107. Parr AM, Tator CH, Keating A. Bone marrow-derived mesenchymal stromal cells for the repair of central nervous system injury. Bone Marrow Transplant 2007;40:609–19. https://doi.org/10.1038/sj.bmt.1705757
  108. Pawluk H, Woźniak A, Grześk G, Kołodziejska R, Kozakiewicz M, Kopkowska E, et al. The role of selected pro-inflammatory cytokines in pathogenesis of ischemic stroke. Clin Interv Aging 2020;15:469–84. https://doi.org/10.2147/CIA.S233909
  109. Kyurkchiev D. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J Stem Cells 2014;6:552. https://doi.org/10.4252/wjsc.v6.i5.552
  110. Glenn JD. Mesenchymal stem cells: Emerging mechanisms of immunomodulation and therapy. World J Stem Cells 2014;6:526. https://doi.org/10.4252/wjsc.v6.i5.526
  111. Haddad R, Saldanha-Araujo F. Mechanisms of T-cell immunosuppression by mesenchymal stromal cells: What do we know so far? Biomed Res Int 2014;2014. https://doi.org/10.1155/2014/216806
  112. Ooi YY, Dheen ST, Sam Wah Tay S. Paracrine effects of mesenchymal stem cells-conditioned medium on microglial cytokines expression and nitric oxide production. Neuroimmunomodulation 2015;22:233–42. https://doi.org/10.1159/000365483
  113. Saldaña L, Vallés G, Bensiamar F, Mancebo FJ, García-Rey E, Vilaboa N. Paracrine interactions between mesenchymal stem cells and macrophages are regulated by 1,25-dihydroxyvitamin D3. Sci Rep 2017;7. https://doi.org/10.1038/s41598-017-15217-8
  114. Kean TJ, Lin P, Caplan AI, Dennis JE. MSCs: Delivery routes and engraftment, cell-targeting strategies, and immune modulation. Stem Cells Int 2013;2013. https://doi.org/10.1155/2013/732742
  115. Caplan H, Olson SD, Kumar A, George M, Prabhakara KS, Wenzel P, et al. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application. Front Immunol 2019;10:1645. https://doi.org/10.3389/fimmu.2019.01645
  116. Matsumoto J, Stewart T, Banks WA, Zhang J. The Transport Mechanism of Extracellular Vesicles at the Blood-Brain Barrier. Curr Pharm Des 2018;23:6206–14. https://doi.org/10.2174/1381612823666170913164738
  117. Bang OY, Kim EH. Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Stroke: Challenges and Progress. Front Neurol 2019;10. https://doi.org/10.3389/fneur.2019.00211
  118. Xin H, Katakowski M, Wang F, Qian JY, Liu XS, Ali MM, et al. MicroRNA-17–9. Cluster in Exosomes Enhance Neuroplasticity and Functional Recovery After Stroke in Rats. Stroke, vol. 48, Lippincott Williams and Wilkins; 2017, p. 747–53. https://doi.org/10.1161/STROKEAHA.116.015204
  119. Asgarpour K, Shojaei Z, Amiri F, Ai J, Mahjoubin-Tehran M, Ghasemi F, et al. Exosomal microRNAs derived from mesenchymal stem cells: Cell-to-cell messages. Cell Commun Signal 2020;18. https://doi.org/10.1186/s12964-020-00650-6
  120. Oggero S, Austin-Williams S, Norling LV. The contrasting role of extracellular vesicles in vascular inflammation and tissue repair. Front Pharmacol 2019;10. https://doi.org/10.3389/fphar.2019.01479
  121. Upadhya R, Madhu LN, Attaluri S, Gitaí DLG, Pinson MR, Kodali M, et al. Extracellular vesicles from human iPSC-derived neural stem cells: miRNA and protein signatures, and anti-inflammatory and neurogenic properties. J Extracell Vesicles 2020;9. https://doi.org/10.1080/20013078.2020.1809064
  122. Hofer HR, Tuan RS. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res Ther 2016;7. https://doi.org/10.1186/s13287-016-0394-0
  123. Maacha S, Sidahmed H, Jacob S, Gentilcore G, Calzone R, Grivel JC, et al. Paracrine Mechanisms of Mesenchymal Stromal Cells in Angiogenesis. Stem Cells Int 2020;2020. https://doi.org/10.1155/2020/4356359
  124. Tsiapalis D, O’Driscoll L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020;9. https://doi.org/10.3390/cells9040991
  125. Nawaz M, Shah N, Zanetti B, Maugeri M, Silvestre R, Fatima F, et al. Extracellular Vesicles and Matrix Remodeling Enzymes: The Emerging Roles in Extracellular Matrix Remodeling, Progression of Diseases and Tissue Repair. Cells 2018;7:167. https://doi.org/10.3390/cells7100167
  126. Candela ME, Geraci F, Turturici G, Taverna S, Albanese I, Sconzo G. Membrane vesicles containing matrix metalloproteinase-9 and fibroblast growth factor-2 are released into the extracellular space from mouse mesoangioblast stem cells. J Cell Physiol 2010;224:144–51. https://doi.org/10.1002/jcp.22111
  127. Xie M, Xiong W, She Z, Wen Z, Abdirahman AS, Wan W, et al. Immunoregulatory Effects of Stem Cell-Derived Extracellular Vesicles on Immune Cells. Front Immunol 2020;11. https://doi.org/10.3389/fimmu.2020.00013
  128. Zhang L, Graf I, Kuang Y, Zheng X, Haupt M, Majid A, et al. Neural progenitor cell-derived extracellular vesicles enhance blood-brain barrier integrity by NF-κB (Nuclear Factor-κB)-Dependent regulation of ABCB1 (ATP-Binding Cassette Transporter B1) in Stroke Mice. Arterioscler Thromb Vasc Biol 2021;41:1127–45. https://doi.org/10.1161/ATVBAHA.120.315031
  129. Morton MC, Neckles VN, Seluzicki CM, Holmberg JC, Feliciano DM. Neonatal Subventricular Zone Neural Stem Cells Release Extracellular Vesicles that Act as a Microglial Morphogen. Cell Rep 2018;23:78–89. https://doi.org/10.1016/j.celrep.2018.03.037
  130. Han Z, Chen F, Ge X, Tan J, Lei P, Zhang J. MiR-21 alleviated apoptosis of cortical neurons through promoting PTEN-Akt signaling pathway in vitro after experimental traumatic brain injury. Brain Res 2014;1582:12–20. https://doi.org/10.1016/j.brainres.2014.07.045
  131. Slota JA, Booth SA. MicroRNAs in neuroinflammation: Implications in disease pathogenesis, biomarker discovery and therapeutic applications. Non-Coding RNA
  132. Seki T, Kanagawa M, Kobayashi K, Kowa H, Yahata N, Maruyama K, et al. Galectin 3- binding protein suppresses amyloid-β production by modulating β-cleavage of amyloid precursor protein. J Biol Chem 2020;295:3678–91. https://doi.org/10.1074/jbc.RA119.008703
  133. Gao W, Li F, Liu L, Xu X, Zhang B, Wu Y, et al. Endothelial colony-forming cell-derived exosomes restore blood-brain barrier continuity in mice subjected to traumatic brain injury. Exp Neurol 2018;307:99–108. https://doi.org/10.1016/j.expneurol.2018.06.001
  134. Davis C, Savitz SI, Satani N. Mesenchymal Stem Cell Derived Extracellular Vesicles for Repairing the Neurovascular Unit after Ischemic Stroke. Cells 2021;10. https://doi.org/10.3390/cells10040767
  135. Öztürk S, Elçin AE, Koca A, Elçin YM. Therapeutic Applications of Stem Cells and Extracellular Vesicles in Emergency Care: Futuristic Perspectives. Stem Cell Rev Reports 2021;17:390–410. https://doi.org/10.1007/s12015-020-10029-2
  136. Petrou P, Gothelf Y, Argov Z, Gotkine M, Levy YS, Kassis I, et al. Safety and Clinical Effects of Mesenchymal Stem Cells Secreting Neurotrophic Factor Transplantation in Patients With Amyotrophic Lateral Sclerosis: Results of Phase 1/2 and 2a Clinical Trials. JAMA Neurol 2016;73:337–44. https://doi.org/10.1001/jamaneurol.2015.4321
  137. Lee JS, Hong JM, Moon GJ, Lee PH, Ahn YH, Bang OY, et al. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells 2010;28:1099–106. https://doi.org/10.1002/stem.430
  138. Levy ML, Crawford JR, Dib N, Verkh L, Tankovich N, Cramer SC. Phase I/II study of safety and preliminary efficacy of intravenous allogeneic mesenchymal stem cells in chronic stroke. Stroke 2019;50:2835–41. https://doi.org/10.1161/STROKEAHA.119.026318
  139. Bhasin A, Srivastava MVP, Kumaran SS, Mohanty S, Bhatia R, Bose S, et al. Autologous Mesenchymal Stem Cells in Chronic Stroke. Cerebrovasc Dis Extra 2011;1:93–104. https://doi.org/10.1159/000333381
  140. Dang XTT, Kavishka JM, Zhang DX, Pirisinu M, Le MTN. Extracellular Vesicles as an Efficient and Versatile System for Drug Delivery. Cells 2020;9. https://doi.org/10.3390/cells9102191
  141. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol Ther 2015;23:812–23. https://doi.org/10.1038/mt.2015.44