Next Article in Journal
Induction of Hepcidin Expression in the Renal Cortex of Sickle Cell Disease Mice
Previous Article in Journal
Vps4a Regulates Autophagic Flux to Prevent Hypertrophic Cardiomyopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanodrugs for the Treatment of Ischemic Stroke: A Systematic Review

1
Doctoral School, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
2
Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul 34214, Turkey
3
Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 115680-8000, USA
4
Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
5
Department of Neurology, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(13), 10802; https://doi.org/10.3390/ijms241310802
Submission received: 1 June 2023 / Revised: 26 June 2023 / Accepted: 26 June 2023 / Published: 28 June 2023
(This article belongs to the Special Issue New Therapeutic Tools to Combat Aging and Cerebrovascular Diseases)

Abstract

:
Ischemic stroke, a significant neurovascular disorder, currently lacks effective restorative medication. However, recently developed nanomedicines bring renewed promise for alleviating ischemia’s effects and facilitating the healing of neurological and physical functions. The aim of this systematic review was to evaluate the efficacy of nanotherapies in animal models of stroke and their potential impact on future stroke therapies. We also assessed the scientific quality of current research focused on nanoparticle-based treatments for ischemic stroke in animal models. We summarized the effectiveness of nanotherapies in these models, considering multiple factors such as their anti-inflammatory, antioxidant, and angiogenetic properties, as well as their safety and biodistribution. We conclude that the application of nanomedicines may reduce infarct size and improve neurological function post-stroke without causing significant organ toxicity.

1. Introduction

Stroke, which accounts for approximately 5.5 million deaths annually, stands as the second leading cause of death worldwide. Beyond the significant mortality rate, stroke also inflicts substantial morbidity, with nearly half of survivors experiencing long-term disabilities. As a result, stroke carries considerable economic and social implications, making it a matter of utmost importance in public health. According to a study conducted by Donkor ES et al. in 2018, it is projected that by 2030, there will be a staggering 23 million stroke cases, resulting in 7.8 million associated deaths [1]. Recent findings from an American Heart Association meeting reveal that ischemic stroke constitutes nearly 87% of all stroke cases [2].
Ischemic stroke, a severe neurological condition, has limited therapeutic options available. Thrombolytic therapy is the sole FDA-approved clinical treatment for ischemic stroke. However, the presence of a physiological barrier complicates the development of effective therapeutic and diagnostic strategies for ischemic stroke [3]. After an ischemic injury, the blood–brain barrier represents a significant obstacle to the penetration of both small- and large-scale therapeutic agents into the brain [4].
After an ischemic stroke, local microglia undergo polarization towards the M1 phenotype due to exposure to proinflammatory cytokines such as IFN-γ and TNF-α. Subsequently, these cells show significant expression of inducible NO (iNOS) to synthesize NO and generate pro-inflammatory cytokines such as TNF, interleukin (IL)-1, and IL-12. The outcome of this M1 polarization event depends on various factors, including the production of iNOS, reactive oxygen species (ROS), and the activation of the inflammasome complex known as NOD-like receptor family pyrin domain-containing 3 (NLRP3), all of which contribute to secondary brain damage [5,6].
The production of anti-inflammatory cytokines (IL-10, IL-13, TGF-β, VEGF, EGF, and Arg1) by M2 microglia has a beneficial impact in cases of ischemia or hypoxia following an ischemic stroke. This production helps to deactivate pro-inflammatory cell types, restore balance within the body, and enhance the outcome of strokes [5,6]. Consequently, modulating the shift from proinflammatory M1 microglia to anti-inflammatory M2 microglia shows promise as a therapeutic strategy for stroke treatment [7].
Nanotechnology holds promising prospects for the treatment of various central nervous system diseases in the field of medical research [8]. Nanoparticles (NPs) are solid colloidal particles, with sizes ranging from 1 to 1000 nm, and they find diverse applications in medicine. Utilizing their small size and mobility, NPs can effectively reach different tissues and cells, enabling both extracellular and intracellular administration. Several materials and techniques can be employed to fabricate nanoparticles, adjusting various parameters to achieve the desired NPs. Key considerations for drug delivery include size, payload encapsulation efficacy, zeta potential, and payload release characteristics [4]. Since the brain’s smallest capillaries have a diameter of only 5–6 μm, administering NPs via microcirculation emerges as a practical approach to facilitate drug delivery to the brain. NPs provide a means to deliver drugs, proteins, vaccines, biological macromolecules, and gene therapy, regardless of their hydrophilic or hydrophobic nature [9].
Nanomaterials offer the potential for molecular and supramolecular interactions with biological systems, which are advantageous. Utilizing these interactions makes it possible to induce appropriate physiological responses in cells while minimizing negative effects on the overall system [10].
In this systematic review, our objective was to evaluate the scientific quality of current research focusing on nanoparticle-based treatments for ischemic stroke. Additionally, we aimed to assess the potential effectiveness of future therapies. This study also discusses the application of nanoparticles for stroke treatment in animal models. We conducted an analysis of their efficacy, taking into account a range of parameters including their anti-inflammatory, antioxidant, and angiogenetic properties, as well as safety and biodistribution.

2. Methods

2.1. Search Strategy

An extensive search was conducted in the PubMed and Web of Science databases using the keywords “nano-drug”, “treatment”, and “stroke” (Supplementary Table S1). To refine the results, we filtered for English language publications, studies on animal species, original work with free access, and articles published within the past 5 years (2018–2022). Duplicate studies were eliminated. The remaining studies were initially assessed based on their abstracts to remove irrelevant records. Subsequently, all the remaining articles were thoroughly read, with only the most relevant ones being included in the study.

2.2. Inclusion and Exclusion Criteria

Only original articles written in English were considered for inclusion. The following inclusion criteria were used: (1) studies that used in vivo models of ischemic brain lesions and (2) studies that used nanoparticles, loaded or not with some type of drug, for stroke treatment. Research involving cell cultures, nano-treatment for animals without stroke lesions, studies using drug therapy on a larger scale than the nanoscale, and studies utilizing nanotechnology for paraclinical exploration were excluded (Supplementary Table S2).
Articles that were indexed in more than one database (duplicates), incomplete articles, abstracts, reviews, and book chapters, as well as studies involving ischemic injury in locations other than the brain were excluded

2.3. Data Extraction and Quality Assessment

Each study provided various details. These included author information, publication year, and the journal in which the study was published. The study’s 2021 impact factor was also listed. Information regarding the animal species used and the number of animals involved was included. Details about the targeted nanoparticles, the potential drug treatment, and the stroke procedure model were provided as well. Other aspects noted were the administration route, treatment time points, and methodological and reporting features. These were assessed through a quality questionnaire designed specifically for pre-clinical studies [11].

3. Results

Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram [12], the search strategy identified 230 potentially related titles. Three duplicate studies were removed, leaving 227 records, of which only 38 studies (16.74%) met the inclusion criteria. Of the remaining 189 PubMed records, 19 studies (10.05%) met the eligibility criteria. Some 55 studies (29.1%) were excluded because they were not conducted on an animal model of ischemic stroke, 20 studies (10.58%) were not pre-clinical trials, 15 (7.94%) were not based on nanomedicine, and 29 (15.34%) did not involve drug therapy. Out of the 89 Web of Science articles, 19 (21.34%) met the inclusion criteria. Some 35 (39.32%) were excluded because they were not conducted on an animal model of ischemic stroke, 16 (17.97%) were not pre-clinical trials, 6 (6.74%) were not based on nanomedicine, 12 (13.48%) did not involve drug therapy, and 1 (1.12%) was not available for full free access (Figure 1).

3.1. Characteristics of the Study

The eligible articles were published between 2018 and 2022. These studies were conducted in various countries, distributed as follows: China (35.84%), the USA (31.57%), England (13.15%), Australia (7.89%), Japan (2.63%), Switzerland (5.26%), and the Republic of Korea (2.63%). The sample size in these studies ranged from 12 to 287 rodents. However, in five studies, the number of animals was not clearly defined. All studies involved animals with ischemic stroke and used the following ischemia models: the MCAO procedure (86.84%), the phototrombotic model (2.63%), the BCCAO procedure (2.63%), the RCCAO procedure (2.63%), and permanent MCAO (2.63%); one study used the first two models of ischemic stroke (2.63%). For the research, studies on rodents with stroke were conducted on mice (55.26%) and rats (42.10%), and one study included both species (2.63%). Six studies were indexed in journals with a 2021 Impact Factor (IF) below 5 (15.79%), twenty-one were indexed in journals with a 2021 IF between 5 and 10 (55.26%), six were in journals with a 2021 IF between 10 and 15 (15.78%), and five were indexed in journals with a 2021 IF above 15 (13.15%). The characteristics of the studies can be found in Table 1.

3.2. Methodological Assessment

The SYRCLE criteria are presented in Supplementary Table S3 and Figure 2. Utilizing SYRCLE’s risk of bias tool, which comprises ten items across six categories of bias, we evaluated the risk of bias in the included studies. These categories included selection, performance, detection, attrition, reporting, and other biases [51]. The term “Yes” indicates a low risk of bias, while “No” signifies a high risk of bias. A “?” symbolizes an unknown risk of bias. Overall, none of the studies included satisfied all the SYRCLE criteria, as the risk from other potential sources of bias remained unclear.
On average, the included studies reported six out of the ten characteristics. The range of scores varied, with the lowest score being 2 out of 10 items (5.12%), and the highest-scoring studies reporting 9 out of 10 items (15.38%). All the studies fulfilled two of the three selection bias criteria, as they described the methods used to generate the allocation sequence in enough detail to evaluate whether it should produce comparable groups, and they used similar groups of animals at baseline. In 64.10% of the included research, the animals were randomly housed during the experiment, and 43.58% of the outcome results were analyzed in a blind manner.

3.3. Nanoparticles and Nanodrugs

The nanoparticles associated with stroke treatment in an animal model are summarized in Table 2. Overall, more than 20 different nanoparticle formulations were discovered, among which the most commonly reported were polylactic-co-glycolic acid (PLGA) (12.82%), exosomes (12.82%), liposomes (10.25%), and extracellular vesicles (5.29%).
Twenty-eight studies (71.79%) administered nanoparticles intravenously via the tail vein; four studies (10.25%) administered them intraperitoneally; one study combined the two; and two studies (5.12%) administered them intrathecally. Additional administration methods used were intranasally, transdermally, and orally.
The animal model of ischemia was treated with nanoparticles in multiple doses in 18 records (46.15%) and in a single dose in the remaining 21 records (53.84%). In 32 records (82.05%), nanoparticles were administered after ischemic or reperfusion surgery, while they were used as a pretreatment in 5 studies (12.82%).

4. Discussion

Ischemic stroke is a severe cerebrovascular disease for which no existing restorative drugs are available. Recently developed nanodrugs offer fresh hope to mitigate the effects of ischemia and stimulate the recovery of neurological and physical functions. Ischemia and reperfusion can trigger a series of events, including oxidative stress and inflammatory responses. These are primarily caused by neutrophils infiltrating the post-stroke brain and by the activation of microglia due to neurological damage. Hence, nanodrugs can limit neutrophil infiltration into the ischemic brain and shift the ratio of M1/M2 phenotypes towards the neuroprotective M2 phenotype. Treatments with nanodrugs also reduce infarct volume and neurological dysfunction, with no notable organ toxicity.

4.1. The Biodistribution of Nanoparticles

The potential for harmful side effects on primary organs significantly hinders the future scope of nanomedicine in biological applications. Several studies have been conducted to investigate the distribution and potential toxicity of nanoparticles in the body. The methodologies employed include histological examination, in vivo imaging, and the measurement of transaminase activity levels, as reported in various reviewed articles.
Research conducted by Lizhen He and his colleagues [37], Lin Guo and her team [30], Daozhou Liu and his group [17], and Chao Li and his associates [11] has involved detailed histological examination of major organs in rats that had suffered a stroke, using H&E staining. The key organ sections examined included those from the heart, liver, spleen, lung, and kidney. No visible inflammation or other pathological changes were observed following exposure of these organs to nanoparticles. This suggests that there was no organ toxicity as a result of nanomedicine administration. The safety of nanomedicine was further corroborated by histological analysis correlated with the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), which were found to be within normal limits [11,17].

4.2. The Antioxidant Activity of Nanoparticles in Stroke Models

Ischemia and reperfusion can trigger a cascade of events, including oxidative stress and an inflammatory response, which may eventually lead to irreversible neuron damage. A major cause of oxidative stress is the excessive production of reactive oxygen species (ROS), which activate apoptotic and necrotic pathways, leading to cell necrosis or death [11].
Ma@(MnO2+FTY), a macrophage-disguised, FTY-loaded MnO2 nanoparticle, was first investigated by Chao Li et al. [11]. Its effectiveness was initially assessed in vitro by measuring the residual H2O2 after incubation with various concentrations of MnO2 for specific time periods. The results showed that MnO2 could effectively scavenge H2O2 and that the scavenging process was dependent on both the concentration of MnO2 and the duration of incubation. When Ma@(MnO2 + FTY) was administered in vivo, it was found to alleviate brain injury and modulate the pro-inflammatory microenvironment [11]. An elevated concentration of reactive oxygen species (ROS) triggers the opening of the mitochondrial permeability transition pore (mPTP) in neurons, subsequently diminishing the potential of the mitochondrial membrane. This initiates a damaging cycle between the mPTP opening and ROS generation. The outbreak of ROS brought about by cerebral ischemia/reperfusion, along with the excessive opening of mPTP in neuronal cells, can be curbed through CsA-loaded HFn nanoparticles (CsA@HFn). These nanoparticles have been shown to mitigate neuronal damage by decreasing the production of ROS [17].
Malondialdehyde (MDA) and superoxide dismutase (SOD) serve as critical benchmarks in the assessment of oxidative stress related to cerebral ischemia/reperfusion damage. An imbalance in oxidative stress during ischemic brain injury leads to a decline in SOD levels and a surge in MDA levels. In the research conducted by Xinyi Yuan and team, the stroke-affected animals that were administered DBZ or DBZ-NLC exhibited a reduction in MDA levels [29]. Following the intrathecal administration of POM nanoclusters, the study led by Shiyong Li and his team revealed that SOD and MDA levels were similar to those found in control groups [34].
Heme oxygenase-1 (HO-1), a crucial antioxidant enzyme, and nuclear factor erythroid-2-related factor 2 (Nrf2), a key component of antioxidant signaling that promotes the transcription of several antioxidant genes and maintains redox equilibrium in cells, were notably up-regulated by treatments with PEG-PTT-T-PEG NP [32] and Que-loaded mAb GAP43-conjugated exosomes [30].
Nanoparticles carrying drugs have been found to enhance the formation of new blood vessels in models of stroke. This is important because, after a stroke, the core area of the brain that is affected by the lack of blood flow experiences damage to its vasculature. The process of angiogenesis, which leads to the development of new blood vessels, is a crucial factor in repairing and remodeling these damaged areas. This process largely depends on the successful reinstatement of blood supply. Consequently, recent research has shifted its focus to enhancing vascularization after a stroke [22].
Prior work by Cheng Wang and colleagues [22] discovered that FE is packed with various growth factors, including BDNF, GDNF, TGF-beta, and bFGF. In their most recent study, they devised a biomimetic nanocarrier that can accurately deliver FE as a treatment for stroke. The peptide sequence Arg-Gly-Asp (RGD) is particularly attractive to the αvβ3 integrin, which is found on the surface of blood vessels undergoing angiogenesis. Meanwhile, PLTs have numerous receptors that can adhere specifically to damaged and inflamed vasculature. The interaction between FE particles coated with RGD-PLT facilitates the sustained release of FE at the stroke site, delivering the neurotrophic factors BDNF, GDNF, and bFGF to the brain. This interaction ultimately results in an increase in blood flow and recovery in neurobehavioral functions [22].
Huixin Zhang and colleagues utilized exosomes to carry miR-210, a substance known to enhance angiogenesis via the VEGF signaling pathway [28]. Taking advantage of the affinity between RGD and αvβ3 integrin—a component found on angiogenic blood vessels—the team linked the exosomes with RGD to transport miR-210 to the site of a lesion. The study confirmed that mice treated with RGD-exo:miR-210 showed an increase in microvessel density within the lesion site, as evidenced by CD34+ per field, at both 7 and 14 days post-treatment, compared to the control groups [28].
In a separate study, Haoan Wu and his team discovered that treatment with glyburide-loaded ASPTT NPs significantly encouraged angiogenesis in the ischemic brain [32]. The researchers found an impressive increase in the number of new blood vessels in the brains of mice receiving this treatment, compared to controls, as confirmed by CD31 immunohistochemistry (IHC) in the ischemic stroke brains.

4.3. The Role of Nanomedicines in Reducing Inflammation in Stroke Models

In the process of injury, inflammation plays a critical role, amplifying the extent of neuronal damage following ischemic injury to the central nervous system. Studies investigating the levels of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β have shown dose-dependent reductions, illustrating the anti-inflammatory effects of nanoparticles [11,13,16,25,31,37,39,41].
Research assessing myeloperoxidase (MPO) activity in the brain—an indicator of neutrophil tissue infiltration—by Zhang and colleagues [15] and Dong and his team [31] has demonstrated that neutrophil infiltration into the ischemic brain can be limited by Doxorubicin and Resolvin D2-loaded nanovehicles.
Research by Koteswara Rao Nalamolu et al. [13] and Tsogzolmaa Ganbold et al. [26] found that Toll-like receptors (TLRs) are instrumental in controlling post-ischemic inflammatory responses. They developed nanoparticles carrying short interference RNAs to target TLRs 2 and 4. The outcome was a substantial reduction in TLR2 and TLR4 gene expression, which reversed cytokine expression patterns and limited the release of numerous proinflammatory cytokines, such as iNOS. This also encouraged the production of several anti-inflammatory cytokines and neurotrophils, leading to an improvement in neurological function. The intricate nature of the signaling systems involved, along with their accompanying inflammatory responses, poses considerable challenges for the development of anti-stroke drugs. Signaling networks play a crucial role in neuronal growth and metabolism in mammalian CNSs, and any disruption to these pathways during or post-stroke can lead to neuronal injury and death [36]. Research by Tian-Tian et al. [10] suggested that the anti-inflammatory action induced by EVReN strongly relies on the MAPK pathway being inhibited by the miRNAs incorporated into EVReN. The findings by Hamed Amani et al. [36] showed that the Jak2 pathway was substantially lowered in the presence of OX26-PEG-Se NPs.
Microglia constitute approximately 10% of all cells in the brain [8]. In the event of an ischemic stroke, these microglia become stimulated and may differentiate into one of two phenotypes: the M1 phenotype, which promotes inflammation, or the M2 phenotype, which mitigates inflammation. M2 microglia assist in recovery following a stroke. Conversely, M1 microglia release a range of pro-inflammatory factors, including IL-1, IL-6, IFN, and TNF, which contribute to additional brain damage. These factors significantly contribute to the worsening of cerebral injury.
In their studies, researchers used Iba-1 immunostaining, a method for tracking the activation of microglia, to observe the response of microglia after being treated with various substances delivered by nanoparticles [10,17,19]. Specifically, in the research conducted by Tian and colleagues [10], microglia exhibited extensive branching of processes after being exposed to extracellular vesicles derived from neural progenitor cells. This was in stark contrast to the microglia in the control group, which maintained thin ramified processes and small cellular bodies.
In an effort to ascertain the ratio of M1 and M2 microglia types within the area of cerebral infarction, research conducted by Daozhou Liu and his colleagues [17], as well as Po-Wah So and his team [19], employed different labeling methods. The M1 type was marked by CD16/32, while CD206 was used to indicate the M2 type. When compared with the control group, a decline in the quantity of cells co-expressing CD16/32 was noted following treatment involving heavy chain ferritin nanoparticles loaded with cyclosporine A and liposomal nanoparticles encapsulated with acetate. Conversely, there was an uptick in the count of cells co-expressing CD206.
Hyo Jung Shin and colleagues analyzed the potential of Perampanel for neuroprotection [16]. The researchers utilized PLGA NPs to enhance the delivery of Perampanel to microglia. Their results indicated that when Perampanel was successfully delivered to microglia, it managed to decrease the emission of pro-inflammatory cytokines while promoting the presence of M2 phenotype microglia, thereby contributing to neuroprotection.

4.4. Treatment with Nanodrugs Reduced the Infarct Volume

The volume of the infarction was determined using triphenyltetrazolium chloride (TTC) staining in 24 out of the total 38 evaluated studies. Compared to the control groups, the group treated with nanoparticles showed a significant reduction in the infarct area, with the reduction scaling up to 80% depending on the dosage [12,13,16,17,18,20,21,23,24,25,26,27,30,31,33,34,35,36,37,40,41,42,43,44,47].

4.5. Impact of Nanotherapy on the Restoration of Neurological Function

The evaluation of neurological recuperation was carried out using behavioral metrics in a significant portion of the included studies (68.42%). Several tests were employed in this regard, including the adhesive removal (or sticky tape) test, the beam walk test, the rotarod performance test, the cylinder test, and the grid walking test, to name a few [12,13,15,16,17,18,21,22,23,24,25,26,27,30,31,32,33,34,35,36,37,38,39,41,42,43,44,45,46,47].
Functional recovery of the nervous system was evaluated by conducting a variety of tests that comprised the components of mNSS, such as motor, sensory, reflex, and balance assessments. These testing methodologies were extensively applied in numerous studies, which utilized the filament stroke model, a method adapted by Longa and his team [49], to gauge the severity of post-stroke damage and the pace of recovery [13,22,34,35,47]. Notably, animal groups that received nanodrug treatments displayed significantly reduced neurological dysfunction, as demonstrated by their lower mNSS scores compared to the control group. This points towards faster recuperation [18,21,25].

5. Conclusions

Ischemic stroke, a significant neurovascular disorder, currently lacks effective restorative medication. However, recently developed nanomedicines bring renewed promise in alleviating ischemia effects and facilitating the healing of neurological and physical functions. The process of ischemia and reperfusion initiates a chain reaction of events, primarily involving oxidative stress and inflammatory responses due to neutrophils infiltrating the brain after a stroke and the activation of microglia resulting from neurological injury. Recently, it has been recognized that the production of anti-inflammatory cytokines by M2 microglia benefits ischemia or hypoxia after an ischemic stroke by deactivating pro-inflammatory cells, restoring balance, and improving stroke outcomes. Therefore, modulating the shift from pro-inflammatory M1 microglia to anti-inflammatory M2 microglia holds promise as a therapeutic approach for stroke treatment. Nanodrugs may have potential benefits in the treatment of cerebral ischemia by restricting neutrophil penetration into the ischemic brain and encouraging a shift in the M1/M2 phenotype ratio in favor of the neuroprotective M2 phenotype. The application of nanodrugs also may reduce infarct size and improve neurological recovery without causing significant organ toxicity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms241310802/s1.

Author Contributions

M.R., A.C. and C.V.A. wrote the main manuscript text. E.K., B.C., A.G., D.M.H. and A.P.-W. reviewed the manuscript. A.P.-W. edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The Article Processing Charges were funded by the University of Medicine and Pharmacy of Craiova, Romania.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

This work was supported by the grant POCU/993/6/13-153178, co-financed by the European Social Fund within the Sectorial Operational Program Human Capital 2014–2020.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Donkor, E.S. Stroke in the21stCentury: A Snapshot of the Burden, Epidemiology, and Quality of Life. Stroke Res. Treat. 2018, 2018, 3238165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Malik, P.; Anwar, A.; Patel, R.; Patel, U. Expansion of the dimensions in the current management of acute ischemic stroke. J. Neurol. 2021, 268, 3185–3202. [Google Scholar] [CrossRef] [PubMed]
  3. Lv, W.; Liu, Y.; Li, S.; Lv, L.; Lu, H.; Xin, H. Advances of nano drug delivery system for the theranostics of ischemic stroke. J. Nanobiotechnol. 2022, 20, 248. [Google Scholar] [CrossRef] [PubMed]
  4. Bharadwaj, V.N.; Nguyen, D.T.; Kodibagkar, V.D.; Stabenfeldt, S.E. Nanoparticle-Based Therapeutics for Brain Injury. Adv. Healthc. Mater. 2018, 7, 1700668. [Google Scholar] [CrossRef]
  5. Orihuela, R.; McPherson, C.A.; Harry, G.J. Microglial M1/M2 polarization and metabolic states. Br. J. Pharmacol. 2016, 173, 649–665. [Google Scholar] [CrossRef] [Green Version]
  6. Jiang, C.T.; Wu, W.F.; Deng, Y.H.; Ge, J.W. Modulators of microglia activation and polarization in ischemic stroke. Mol. Med. Rep. 2020, 21, 2006–2018. [Google Scholar] [CrossRef] [Green Version]
  7. Mao, J.-H.; Xu, Y.; Li, B.-W.; Yang, Y.-L.; Peng, Y.; Zhi, F. Microglia polarization in ischemic stroke: Complex mechanisms and therapeutic interventions. Chin. Med. J. 2021, 134, 2415–2417. [Google Scholar] [CrossRef]
  8. Alkaff, S.A.; Radhakrishnan, K.; Nedumaran, A.M.; Liao, P.; Czarny, B. Nanocarriers for Stroke Therapy: Advances and Obstacles in Translating Animal Studies. Int. J. Nanomed. 2020, 15, 445–464. [Google Scholar] [CrossRef] [Green Version]
  9. Hans, M.; Lowman, A. Biodegradable nanoparticles for drug delivery and targeting. Curr. Opin. Solid State Mater. Sci. 2002, 6, 319–327. [Google Scholar] [CrossRef]
  10. Gilmore, J.L.; Yi, X.; Quan, L.; Kabanov, A.V. Novel nanomaterials for clinical neuroscience. J. NeuroImmune Pharmacol. 2008, 3, 83. [Google Scholar] [CrossRef] [Green Version]
  11. Vahidy, F.; Schäbitz, W.R.; Fisher, M.; Aronowski, J. Reporting Standards for Preclinical Studies of Stroke Therapy. Stroke 2016, 47, 2435–2438. [Google Scholar] [CrossRef] [Green Version]
  12. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gotzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA Statement for Reporting Systematic Reviews and Meta-Analyses of Studies That Evaluate Healthcare Interventions: Explanation and Elaboration. BMJ 2009, 339, b2700. [Google Scholar] [CrossRef] [Green Version]
  13. Tian, T.; Cao, L.; He, C.; Ye, Q.; Liang, R.; You, W.; Zhang, H.; Wu, J.; Ye, J.; Tannous, B.A.; et al. Targeted delivery of neural progenitor cell-derived extracellular vesicles for anti-inflammation after cerebral ischemia. Theranostics 2021, 11, 6507–6521. [Google Scholar] [CrossRef]
  14. Li, C.; Zhao, Z.; Luo, Y.; Ning, T.; Liu, P.; Chen, Q.; Chu, Y.; Guo, Q.; Zhang, Y.; Zhou, W.; et al. Macrophage-Disguised Manganese Dioxide Nanoparticles for Neuroprotection by Reducing Oxidative Stress and Modulating Inflammatory Microenvironment in Acute Ischemic Stroke. Adv. Sci. 2021, 8, e2101526. [Google Scholar] [CrossRef]
  15. Deng, G.; Ma, C.; Zhao, H.; Zhang, S.; Liu, J.; Liu, F.; Chen, Z.; Chen, A.T.; Yang, X.; Avery, J.; et al. Anti-edema and antioxidant combination therapy for ischemic stroke via glyburide-loaded betulinic acid nanoparticles. Theranostics 2019, 9, 6991–7002. [Google Scholar] [CrossRef]
  16. Nalamolu, K.R.; Challa, S.R.; Fornal, C.A.; Grudzien, N.A.; Jorgenson, L.C.; Choudry, M.M.; Smith, N.J.; Palmer, C.J.; Pinson, D.M.; Klopfenstein, J.D.; et al. Attenuation of the Induction of TLRs 2 and 4 Mitigates Inflammation and Promotes Neurological Recovery After Focal Cerebral Ischemia. Transl. Stroke Res. 2021, 12, 923–936. [Google Scholar] [CrossRef]
  17. Ahmad, S.; Truran, S.; Karamanova, N.; Kindelin, A.; Lozoya, M.; Weissig, V.; Emerson, H.; Griffiths, D.R.; Vail, T.; Lifshitz, J.; et al. Nanoliposomes Reduce Stroke Injury Following Middle Cerebral Artery Occlusion in Mice. Stroke 2022, 53, e37–e41. [Google Scholar] [CrossRef]
  18. Zhang, C.Y.; Dong, X.; Gao, J.; Lin, W.; Liu, Z.; Wang, Z. Nanoparticle-induced neutrophil apoptosis increases survival in sepsis and alleviates neurological damage in stroke. Sci. Adv. 2019, 5, eaax7964. [Google Scholar] [CrossRef] [Green Version]
  19. Shin, H.J.; Lee, K.Y.; Kang, J.W.; Choi, S.G.; Kim, D.W.; Yi, Y.Y. Perampanel Reduces Brain Damage via Induction of M2 Microglia in a Neonatal Rat Stroke Model. Int. J. Nanomed. 2022, 17, 2791–2804. [Google Scholar] [CrossRef]
  20. Liu, D.; Ji, Q.; Cheng, Y.; Liu, M.; Zhang, B.; Mei, Q.; Huan, M.; Zhou, S. Cyclosporine A loaded brain targeting nanoparticle to treat cerebral ischemia/reperfusion injury in mice. J. Nanobiotechnol. 2022, 20, 256. [Google Scholar] [CrossRef]
  21. Song, W.; Bai, L.; Yang, Y.; Wang, Y.; Xu, P.; Zhao, Y.; Zhou, X.; Li, X.; Xue, M. Long-Circulation and Brain Targeted Isoliquiritigenin Micelle Nanoparticles: Formation, Characterization, Tissue Distribution, Pharmacokinetics and Effects for Ischemic Stroke. Int. J. Nanomed. 2022, 17, 3655–3670. [Google Scholar] [CrossRef] [PubMed]
  22. So, P.-W.; Ekonomou, A.; Galley, K.; Brody, L.; Sahuri-Arisoylu, M.; Rattray, I.; Cash, D.; Bell, J.D. Intraperitoneal delivery of acetate-encapsulated liposomal nanoparticles for neuroprotection of the penumbra in a rat model of ischemic stroke. Int. J. Nanomed. 2019, 14, 1979–1991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Otake, H.; Yamaguchi, M.; Ogata, F.; Deguchi, S.; Yamamoto, N.; Sasaki, H.; Kawasaki, N.; Nagai, N. Transdermal System Based on Solid Cilostazol Nanoparticles Attenuates Ischemia/Reperfusion-Induced Brain Injury in Mice. Nanomaterials 2021, 11, 1009. [Google Scholar] [CrossRef] [PubMed]
  24. Duan, R.; Sun, K.; Fang, F.; Wang, N.; He, R.; Gao, Y.; Jing, L.; Li, Y.; Gong, Z.; Yao, Y.; et al. An ischemia-homing bioengineered nano-scavenger for specifically alleviating multiple pathogeneses in ischemic stroke. J. Nanobiotechnol. 2022, 20, 397. [Google Scholar] [CrossRef]
  25. Wang, C.; Yang, X.; Jiang, Y.; Qi, L.; Zhuge, D.; Xu, T.; Guo, Y.; Deng, M.; Zhang, W.; Tian, D.; et al. Targeted delivery of fat extract by platelet membrane-cloaked nanocarriers for the treatment of ischemic stroke. J. Nanobiotechnol. 2022, 20, 249. [Google Scholar] [CrossRef]
  26. Wang, Y.; Wang, Y.; Li, S.; Cui, Y.; Liang, X.; Shan, J.; Gu, W.; Qiu, J.; Li, Y.; Wang, G. Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke. J. Nanobiotechnol. 2021, 19, 331. [Google Scholar] [CrossRef]
  27. Guo, L.; Pan, J.; Li, F.; Zhao, L.; Shi, Y. A novel brain targeted plasma exosomes enhance the neuroprotective efficacy of edaravone in ischemic stroke. IET Nanobiotechnol. 2021, 15, 107–116. [Google Scholar] [CrossRef]
  28. Zhang, Y.; Wang, S.; Lu, F.; Zhang, M.; Kong, H.; Cheng, J.; Luo, J.; Zhao, Y.; Qu, H. The neuroprotective effect of pretreatment with carbon dots from Crinis Carbonisatus (carbonized human hair) against cerebral ischemia reperfusion injury. J. Nanobiotechnol. 2021, 19, 257. [Google Scholar] [CrossRef]
  29. Ganbold, T.; Bao, Q.; Xiao, H.; Zurgaanjin, D.; Liu, C.; Han, S.; Hasi, A.; Baigude, H. Peptidomimetic Lipid-Nanoparticle-Mediated Knockdown of TLR4 in CNS Protects against Cerebral Ischemia/Reperfusion Injury in Mice. Nanomaterials 2022, 12, 2072. [Google Scholar] [CrossRef]
  30. Liu, W.; Su, C.; Qi, Y.; Liang, J.; Zhao, L.; Shi, Y. Brain-targeted heptapeptide-loaded exosomes attenuated ischemia–reperfusion injury by promoting the transfer of healthy mitochondria from astrocytes to neurons. J. Nanobiotechnol. 2022, 20, 242. [Google Scholar] [CrossRef]
  31. Zhang, H.; Wu, J.; Wu, J.; Fan, Q.; Zhou, J.; Wu, J.; Liu, S.; Zang, J.; Ye, J.; Xiao, M.; et al. Exosome-mediated targeted delivery of miR-210 for angiogenic therapy after cerebral ischemia in mice. J. Nanobiotechnol. 2019, 17, 29. [Google Scholar] [CrossRef] [Green Version]
  32. Yuan, X.; Fei, F.; Sun, H.; Xiao, C.; Zhao, X.; Zhang, Y.; Zheng, X. Tanshinol borneol ester on nanostructured lipid carriers has longer brain and systemic effector retention and better antioxidant activity in vivo. Int. J. Nanomed. 2018, 13, 2265–2274. [Google Scholar] [CrossRef] [Green Version]
  33. Guo, L.; Huang, Z.; Huang, L.; Liang, J.; Wang, P.; Zhao, L.; Shi, Y. Surface-modified engineered exosomes attenuated cerebral ischemia/reperfusion injury by targeting the delivery of quercetin towards impaired neurons. J. Nanobiotechnol. 2021, 19, 141. [Google Scholar] [CrossRef]
  34. Dong, X.; Gao, J.; Zhang, C.Y.; Hayworth, C.; Frank, M.; Wang, Z. Neutrophil Membrane-Derived Nanovesicles Alleviate Inflammation To Protect Mouse Brain Injury from Ischemic Stroke. ACS Nano 2019, 13, 1272–1283. [Google Scholar] [CrossRef]
  35. Wu, H.; Peng, B.; Mohammed, F.S.; Gao, X.; Qin, Z.; Sheth, K.N.; Zhou, J.; Jiang, Z. Brain Targeting, Antioxidant Polymeric Nanoparticles for Stroke Drug Delivery and Therapy. Small 2022, 18, e2107126. [Google Scholar] [CrossRef]
  36. Yang, X.; Xu, L.; Zhou, J.; Ge, Y.; Wu, S.; Huang, J.; Li, Y.; Zhu, M.; Jin, X.; Yang, L. Integration of phospholipid-complex nanocarrier assembly with endogenous N-oleoylethanolamine for efficient stroke therapy. J. Nanobiotechnol. 2019, 17, 8. [Google Scholar] [CrossRef]
  37. Li, S.; Jiang, D.; Ehlerding, E.B.; Rosenkrans, Z.T.; Engle, J.W.; Wang, Y.; Liu, H.; Ni, D.; Cai, W. Intrathecal Administration of Nanoclusters for Protecting Neurons against Oxidative Stress in Cerebral Ischemia/Reperfusion Injury. ACS Nano 2019, 13, 13382–13389. [Google Scholar] [CrossRef]
  38. Zhang, D.; Cai, G.; Liu, K.; Zhuang, Z.; Jia, K.; Pei, S.; Wang, X.; Wang, H.; Xu, S.; Cui, C.; et al. Microglia exosomal miRNA-137 attenuates ischemic brain injury through targeting Notch1. Aging 2021, 13, 4079–4095. [Google Scholar] [CrossRef]
  39. Amani, H.; Habibey, R.; Shokri, F.; Hajmiresmail, S.J.; Akhavan, O.; Mashaghi, A.; Pazoki-Toroudi, H. Selenium nanoparticles for targeted stroke therapy through modulation of inflammatory and metabolic signaling. Sci. Rep. 2019, 9, 6044. [Google Scholar] [CrossRef] [Green Version]
  40. He, L.; Huang, G.; Liu, H.; Sang, C.; Liu, X.; Chen, T. Highly bioactive zeolitic imidazolate framework-8–capped nanotherapeutics for efficient reversal of reperfusion-induced injury in ischemic stroke. Sci. Adv. 2020, 6, eaay9751. [Google Scholar] [CrossRef] [Green Version]
  41. Zhang, Q.; Zhu, W.; Xu, F.; Dai, X.; Shi, L.; Cai, W.; Mu, H.; Hitchens, T.K.; Foley, L.M.; Liu, X.; et al. The interleukin-4/PPARγ signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury. PLoS Biol. 2019, 17, e3000330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Azadi, R.; Mousavi, S.E.; Kazemi, N.M.; Yousefi-Manesh, H.; Rezayat, S.M.; Jaafari, M.R. Anti-inflammatory efficacy of Berberine Nanomicelle for improvement of cerebral ischemia: Formulation, characterization and evaluation in bilateral common carotid artery occlusion rat model. BMC Pharmacol. Toxicol. 2021, 22, 54. [Google Scholar] [CrossRef]
  43. Yang, C.; Zhao, Q.; Yang, S.; Wang, L.; Xu, X.; Li, L.; Al-Jamal, W.T. Intravenous Administration of Scutellarin Nanoparticles Augments the Protective Effect against Cerebral Ischemia–Reperfusion Injury in Rats. Mol. Pharm. 2022, 19, 1410–1421. [Google Scholar] [CrossRef]
  44. Luo, L.; Zang, G.; Liu, B.; Qin, X.; Zhang, Y.; Chen, Y.; Zhang, H.; Wu, W.; Wang, G. Bioengineering CXCR4-overexpressing cell membrane functionalized ROS-responsive nanotherapeutics for targeting cerebral ischemia-reperfusion injury. Theranostics 2021, 11, 8043–8056. [Google Scholar] [CrossRef] [PubMed]
  45. Okahara, A.; Koga, J.-I.; Matoba, T.; Fujiwara, M.; Tokutome, M.; Ikeda, G.; Nakano, K.; Tachibana, M.; Ago, T.; Kitazono, T.; et al. Simultaneous targeting of mitochondria and monocytes enhances neuroprotection against ischemia-reperfusion injury. Sci. Rep. 2020, 10, 14435. [Google Scholar] [CrossRef] [PubMed]
  46. Takagishi, S.; Arimura, K.; Murata, M.; Iwaki, K.; Okuda, T.; Ido, K.; Nishimura, A.; Narahara, S.; Kawano, T.; Iihara, K. Protein Nanoparticles Modified with PDGF-B as a Novel Therapy After Acute Cerebral Infarction. Eneuro 2021, 8, 1–14. [Google Scholar] [CrossRef]
  47. Dhuri, K.; Vyas, R.N.; Blumenfeld, L.; Verma, R.; Bahal, R. Nanoparticle Delivered Anti-miR-141-3p for Stroke Therapy. Cells 2021, 10, 1011. [Google Scholar] [CrossRef]
  48. Huang, W.; Wang, L.; Zou, Y.; Ding, X.; Geng, X.; Li, J.; Zhao, H.; Qi, R.; Li, S. Preparation of gastrodin-modified dendrimer-entrapped gold nanoparticles as a drug delivery system for cerebral ischemia-reperfusion injury. Brain Behav. 2022, 12, e2810. [Google Scholar] [CrossRef]
  49. Estevez, A.Y.; Ganesana, M.; Trentini, J.F.; Olson, J.E.; Li, G.; Boateng, Y.O.; Lipps, J.M.; Yablonski, S.E.R.; Donnelly, W.T.; Leiter, J.C.; et al. Antioxidant Enzyme-Mimetic Activity and Neuroprotective Effects of Cerium Oxide Nanoparticles Stabilized with Various Ratios of Citric Acid and EDTA. Biomolecules 2019, 9, 562. [Google Scholar] [CrossRef] [Green Version]
  50. Wang, J.; Zhang, Y.; Xia, J.; Cai, T.; Du, J.; Chen, J.; Li, P.; Shen, Y.; Zhang, A.; Fu, B.; et al. Neuronal PirB Upregulated in Cerebral Ischemia Acts as an Attractive Theranostic Target for Ischemic Stroke. J. Am. Heart Assoc. 2018, 7, e007197. [Google Scholar] [CrossRef] [Green Version]
  51. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.M.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s Risk of Bias Tool for Animal Studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow diagram of the literature search. Note: Adapted from “The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration”, by Liberati et al., 2009 [12].
Figure 1. Flow diagram of the literature search. Note: Adapted from “The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration”, by Liberati et al., 2009 [12].
Ijms 24 10802 g001
Figure 2. Quality study assessed by SYRCLE criteria (Supplementary Table S3).
Figure 2. Quality study assessed by SYRCLE criteria (Supplementary Table S3).
Ijms 24 10802 g002
Table 1. Characteristics of the study.
Table 1. Characteristics of the study.
NrFirst
Author
YearLinkCountryJournalImpact Factor (2021)SpeciesAnimals NumberStroke Model
1Tian T [13]2021https://pubmed.ncbi.nlm.nih.gov/33995671/ (accessed on 1 November 2022)AustraliaTheranostics11.556MouseNKMCAO
2Li C [14]2021https://pubmed.ncbi.nlm.nih.gov/34436822/ (accessed on 1 November 2022)ChinaAdvanced Science17.521Rat15MCAO
3Deng G [15]2019https://pubmed.ncbi.nlm.nih.gov/31660082/ (accessed on 1 November 2022)AustraliaTheranostics11.556Mouse20MCAO
4Nalamolu KR [16]2022https://pubmed.ncbi.nlm.nih.gov/33426628/ (accessed on 1 November 2022) United StatesTranslational Stroke Research6.8Rat77MCAO
5Ahmad S [17]2022https://pubmed.ncbi.nlm.nih.gov/34743535/ (accessed on 1 November 2022)United StatesStroke10.170Mouse15MCAO
6Zhang CY [18]2019https://pubmed.ncbi.nlm.nih.gov/31723603/ (accessed on 1 November 2022)United StatesScience Advances14.136Mouse12MCAO
7Shin HJ [19]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000820480900001 (accessed on 1 November 2022)Republic of KoreaInternational Journal of Nanomedicine7.033Rat19Photothrombotic
8Liu D [20]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000805834400009 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Mouse18MCAO
9Song W [21]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000855167100001 (accessed on 1 November 2022)ChinaInternational Journal of Nanomedicine7.033Mouse24MCAO
10So PW [22]2019https://www.webofscience.com/wos/woscc/full-record/WOS:000462306500001 (accessed on 1 November 2022)EnglandInternational Journal of Nanomedicine7.033Rat20MCAO
11Otake H [23]2021https://www.webofscience.com/wos/woscc/full-record/WOS:000643359300001 (accessed on 1 November 2022)JapanNanomaterials5.719Mouse41MCAO
12Duan R [24]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000849362300001 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Rat50MCAO
13Wang C [25]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000804027800002 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Mouse51Transient MCAO
14Wang Y [26]2021https://www.webofscience.com/wos/woscc/full-record/WOS:000709809800001 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Rat25Transient MCAO
15Guo L [27]2021https://www.webofscience.com/wos/woscc/full-record/WOS:000614878500001 (accessed on 1 November 2022)ChinaIET Nanobiotechnology2.05Rat15Permanent MCAOn
16Zhang Y [28]2021https://www.webofscience.com/wos/woscc/full-record/WOS:000690907800003 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Rat90MCAO
17Ganbold T [29]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000816041900001 (accessed on 1 November 2022)ChinaNanomaterials5.719Mouse24Transient MCAO
18Liu W [30]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000800947800003 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Rat18Transient MCAO
19Zhang H [31]2019https://www.webofscience.com/wos/woscc/full-record/WOS:000459387900001 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429MouseNKTransient MCAO
20Yuan X [32]2018https://www.webofscience.com/wos/woscc/full-record/WOS:000430006300002 (accessed on 1 November 2022)ChinaInternational Journal of Nanomedicine7.033Mouse60Transient MCAO
21Guo L [33]2021https://www.webofscience.com/wos/woscc/full-record/WOS:000655044700003 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Rat18Transient MCAO
22Dong X [34]2019https://www.webofscience.com/wos/woscc/full-record/WOS:000460199400031 (accessed on 1 November 2022)United StatesACS Nano18.027MouseNKMCAO
23Wu H [35]2022https://www.webofscience.com/wos/woscc/full-record/WOS:000770792800001 (accessed on 1 November 2022)United StatesSmall15.153Mouse40MCAO
24Yang X [36]2019https://www.webofscience.com/wos/woscc/full-record/WOS:000456253200002 (accessed on 1 November 2022)ChinaJournal of Nanobiotechnology9.429Rat Mouse38 ratsTransient MCAO
25Li S [37]2019https://www.webofscience.com/wos/woscc/full-record/WOS:000500650000111 (accessed on 1 November 2022)United StatesACS Nano18.027Rat20MCAO
26Zhang D [38]2021https://pubmed.ncbi.nlm.nih.gov/33461167/ (accessed on 1 November 2022)United StatesAging (Albany NY)5.955Mouse36MCAO
27Amani H [39]2019https://pubmed.ncbi.nlm.nih.gov/30988361/ (accessed on 1 November 2022)EnglandScientific Reports4.996Rat60MCAO
28He L [40]2020https://pubmed.ncbi.nlm.nih.gov/32206718/ (accessed on 1 November 2022)United StatesScience Advances14.136Mouse36MCAO
29Zhang Q [41]2019https://pubmed.ncbi.nlm.nih.gov/31226122/ (accessed on 1 November 2022)United StatesPLoS Biology9.593Mouse287MCAO
30Azadi R [42]2021https://pubmed.ncbi.nlm.nih.gov/34600570/ (accessed on 1 November 2022)EnglandBMC Pharmacology and Toxicology2.44Rat96BCCAO
31Yang C [43]2022https://pubmed.ncbi.nlm.nih.gov/35441510/ (accessed on 1 November 2022)United StatesMolecular Pharmaceutics5.364Rat12MCAO
32Luo L [44]2021https://pubmed.ncbi.nlm.nih.gov/34335979/ (accessed on 1 November 2022)AustraliaTheranostics11.556Mouse18MCAO
33Okahara A [45]2020https://pubmed.ncbi.nlm.nih.gov/32879367/ (accessed on 1 November 2022)EnglandScientific Reports4.996MouseNKMCAO
34Takagishi S [46]2021https://pubmed.ncbi.nlm.nih.gov/34462309/ (accessed on 1 November 2022)United StateseNeuro4.363Mouse176Transient MCAO
35Dhuri K [47]2021https://pubmed.ncbi.nlm.nih.gov/33922958/ (accessed on 1 November 2022)SwitzerlandCells6.70Mouse26Transient MCAO
36Huang W [48]2022https://pubmed.ncbi.nlm.nih.gov/36408880/ (accessed on 1 November 2022)United StatesBrain and Behavior19.227Rat25MCAO
37Estevez AY [49]2019https://pubmed.ncbi.nlm.nih.gov/31623336/ (accessed on 1 November 2022)SwitzerlandBiomolecules3.38Rat16RCCAO
38Wang J [50]2018https://pubmed.ncbi.nlm.nih.gov/29378731/(accessed on 1 November 2022) EnglandJournal of the American Heart Association5.501Mouse43MCAO Photothrombotic
Abbreviations: NK, not known; MCAO, Middle Cerebral Artery Occlusion; BCCAO, Bilateral Common Carotid Artery Occlusion; RCCAO, Right Common Carotid Artery Occlusion.
Table 2. Nanoparticles associated with stroke treatment in animal models.
Table 2. Nanoparticles associated with stroke treatment in animal models.
NrFirst AuthorYearSpeciesNanoparticleCombined withRouteTimepointResults
1Tian T [13]2021MouseNeural progenitor cell-derived extracellular vesiclesArginine-glycine -aspartic acid (RGD)-4C peptideIV12 h after MCAO/R- Anti-inflammatory activity
- Significantly strong suppression of TNF-α, IL-1β, and IL-6
- Microglia activation was assessed
2Li C [14]2021RatManganese dioxide nanosphereFingolimodIVAfter a 2 h reperfusion- Good ischemic region targeting ability
- Reducing oxidative stress
- Regulating inflammatory microenvironment
- Reducing behavioral defects
3Deng G [15]2019MouseBetulinic acid extracted from E. ulmoidesGlyburideIV0 h, 24 h, and 48 h after MCAO- Reduced infarct volume, brain edema, and BBB leakage
- Improved mouse survival
- Improved neurological scores
4Nalamolu KR [16]2022RatRNA plasmids formulated as nanoparticles by in vivo-jetPEITLR2shRNA+
TLR4shRNA
IV30 min after reperfusion- Attenuated post-ischemic inflammation
- Preserved motor function, and promoted recovery of the sensory and motor functions
- Decreased infarct volume
5Ahmad S [17]2022MouseNanoliposomes-Intraperitoneally; IVFirst one intraperitoneally 1 h before occlusion and a second dose intravenously immediately before reperfusion- Improved neurological impairment
- Smaller infarcts
- Decreased brain edema
6Zhang CY [18]2019MouseBovine serum albuminDoxorubicinIV1 h after reperfusion- Reduced neutrophil numbers and cytokines in the brain
- Enhanced mouse neurological
recovery
7Shin HJ [19]2022RatPolylactic-co-glycolic acid NPsPerampanelIntrathecally0 h after Rose Bengal-induced photothrombosis- Decreased pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, COX2, and iNOS)
- Increased M2 polarization
- Improved performance in the
hanging test
- Decreased infarct volume
8Liu D [20]2022MouseHeavy chain ferritin nanoparticlesCyclosporine AIV0 h- Selectively delivered to cerebral infarction area
- Decreased infarct volume
- Improved neurological impairment; the number of astrocyte and microglia in cerebral infarction tissue was significantly decreased
- Increased M2 polarization
- Decreased ROS level
- Attenuated the damage of BBB
9Song W [21]2022MouseNanomicelles made of DSPE-PEG2000IsoliquiritigeninIV7 days treatment after MCAO- Improved neurological impairment
- Inhibition of Apoptosis and Autophagy
10So PW [22]2019RatLiposomeAcetateIntraperitoneally0 h and daily for 2 weeks- Decreased infarct volume
- Improved neurological impairment
- Increased M2 polarization
11Otake H [23]2021MouseSolid nanoparticlesCilostazol carbopol gelTransdermallyThree days after reperfusion was administrated once a day and maintained for 66 h- Decreased infarct volume
12Duan R [24]2022RatTannic acid nanoparticle covered with a M2-type microglia membraneCatalaseIV0.5 h after occlusion- Decreased infarct volume
- Increased M2 polarization
- Improved neurological impairment
13Wang C [25]2022MousePolylactic-co- glycolic acid core enclosed by RGD peptide-decorated plasma membrane of PLTsHuman fat extractIVOne day after tMCAO- Targeted damaged and inflamed blood vessels
- Rapid accumulation in the lesion area of ischemic brain
- Delivered BDNF, bFGF, and GDNF to mice brain after stroke, which could contribute to angiogenesis and neurogenesis
- Improved neurobehavioral recovery
14Wang Y [26]2021RatMonocyte membranes-coated rapamycin nanoparticles-IV12 h after reperfusion- Decreased infarct volume
- Improved neurobehavioral recovery
15Guo L [27]2021RatExosomesEndaravoneIVDaily for 7 days- Decreased infarct volume
- Improved neurobehavioral recovery
- Induced the highest number of neurons, as well as evident improvement in the integrity of basic neuronal structure
16Zhang Y [28]2021RatCarbon dotsCrinis
Carbonisatus
IntraperitoneallyPretreatment 6 h and 1 h prior to MCAO- Decreased infarct volume
- Decreased BBB leakage
- Improved neurobehavioral recovery
- Decreased level of TNF-α and IL-6 inhibited excitatory neurotransmitters aspartate and glutamate, and increased the level of 5-hydroxytryptamine
17Ganbold T [29]2022MouseDoGo310 Lipid NPsShort-interference
TLR4
IV3 days after surgery- Promoted the expression of anti-inflammatory factors
- Inhibited proinflammatory cytokines
- Recovery of neurological functions
18Liu W [30]2022RatMacrophage-derived exosomesHeptapeptideIV0 h after reperfusion- Decreased infarct volume
- Improved neurobehavioral recovery
- Improved the mitochondrial function of astrocytes and contributed to the transfer of their healthy mitochondria to nearby damaged neurons, which in turn led to increased neuronal viability and neuroprotection against IS
19Zhang H [31]2019MouseRGD-ExosomemiR-210IVOnce every other day for 14 days, after stroke- Promoted VEGF expression and angiogenesis
- Decreased the level of significant decrease in animal survival
20Yuan X [32]2018MouseNanostructured lipid carriersTanshiol borneol ester(DBZ)-PEGIV0 h after reperfusion- Reduced oxidative stress
21Guo L [33]2021RatmAb GAP43- ExosomesQuercetinIV0 h after reperfusion- Decreased infarct volume
- Improved neurological function
- Reduced ROS production
22Dong X [34]2019MouseNeutrophil membrane- derived nanovesiclesResolvin D2IV1 h after reperfusion- Improved neurological function
- Decreased the level of TNF-α, IL-6, and IL-1β
23Wu H [35]2022MouseAMD3100- conjugated, shrinkable poly (2,2′-thiodiethylene 3,3′-thiodipropionate)GlyburideIV0 h, 24 h, and 48 h after MCAO- Antioxidant effect
- Reduced brain edema
- Repaired compromised BBB
- Promoted the growth of new vessels
- Improved neurological function
24Yang X [36]2019Rat
Mouse
Soybean phosphatidylcholineN-oleoylethanolamineIntragastricallyRats: once daily/14 days after ischemia; mice: once at the time of redispersion- Decreased infarct volume
- Improved neurological function
- Reduced inflammation
25Li S [37]2019RatPolyoxometalae nanoclusters-Intrathecally1 h after MCAO- Antioxidant effects
- Decreased the level of TNF-α and IL-6
- Decreased infarct volume
- Reduced brain edema
- Improved neurological function
26Zhang D [38]2021MouseExosomes derived from microglia in M2 phenotype (BV2-Exo)Reduced expression of miRNA-137 plus CrenIVImmediately after surgery and lasted for three days- Improved neurological function
- Decreased infarct volume
- Decreased apoptosis
27Amani H [39]2019RatSelenium nanoparticlesAnti-transferrin receptor monoclonal antibody (OX26)-PEGylatedIntraperitoneally1 h before ischemic stroke- Attenuated inflammatory
reactions
- Decreased brain edema
- Decreased infarct volume
- Increased neuronal cell survival
- Recovery of locomotor function
28He L [40]2020MouseCeria nanoparticlesZeolitic imidazolate framework-8IVImmediately after reperfusion and injected every other day for 3 days- Decreased infarct volume
- Improved neurological function
- Antioxidant effects
- Decreased the apoptosis of neurons
- Decreased the level of TNF-α, IL-6, and IL-1β
29Zhang Q [41]2019MouseLiposomeIL-14Intranasally6 h after MCAO and repeated at 1–7 d, 14 d, 21 d, and 28 d after MCAO- Improved neurological function
- Promoted white matter integrity
- Improved the structural and functional integrity of myelinated fibers
30Azadi R [42]2021RatNano micellesBerberineOrallyPretreated with the drug (100 mg/kg) and nano-drug (25, 50, 75, and 100 mg/kg) for 14 days- Decreased levels of inflammatory factors TNF-α, IL-1β, and MDA
31Yang C [43]2022RatPLGA nanoparticlesScutellarinIVAfter MCAO once a day for 3 days- Decreased infarct volume
- Improved neurological function
- Decreased the apoptosis of neurons
32Luo L [44]2021MouseBioengineered CXCR4-overexpressed cell membrane coated nanoparticles HOPRapamycinIV0 h after reperfusion- Decreased infarct volume
- Improved neurological function
- Decreased the level of TNF-α and IL-6
- Increased M2 polarization
33Okahara A [45]2022MousePLGA-NPsCyclosporine A
and
pitavastatin
IV0 h after reperfusion- Decreased infarct volume
- Improved neurological function
34Takagishi S [46]2021MouseHeat shock protein NPsPlatelet-derived growth factorIV1 day after surgery- Decreased infarct volume
- Improved neurological function
- Upregulated astrogliosis in peri-infarct area
- Reduced apoptosis in peri-infarct area
35Dhuri K [47]2021MousePLGA-NPsPNA and PS anti-miR-141-3pIV4 h after stroke- Decreased infarct volume
- Decreased the level of TNF-α
- Superior efficacy of PNA-NPs as compared to PS-NPs for miR-141-3p inhibitory activity
36Huang W [48]2022RatPoly(amide amine) (PAMAM) dendrimersGastrodinIVThree times within 7 days- Decrease in apoptosis level
37Estevez AY [49]2019RatCerium oxide nanoparticlesCitric acid and EDTAIntraperitoneally72 h before the surgery- Antioxidant effects
38Wang J [50]2018MouseLiposomesPaired immunoglobulin-like receptor BIVEvery day by tail vein after ischemia until day 7- Decreased infarct volume
- Improved neurological function
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ruscu, M.; Cercel, A.; Kilic, E.; Catalin, B.; Gresita, A.; Hermann, D.M.; Albu, C.V.; Popa-Wagner, A. Nanodrugs for the Treatment of Ischemic Stroke: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 10802. https://doi.org/10.3390/ijms241310802

AMA Style

Ruscu M, Cercel A, Kilic E, Catalin B, Gresita A, Hermann DM, Albu CV, Popa-Wagner A. Nanodrugs for the Treatment of Ischemic Stroke: A Systematic Review. International Journal of Molecular Sciences. 2023; 24(13):10802. https://doi.org/10.3390/ijms241310802

Chicago/Turabian Style

Ruscu, Mihai, Andreea Cercel, Ertugrul Kilic, Bogdan Catalin, Andrei Gresita, Dirk M. Hermann, Carmen Valeria Albu, and Aurel Popa-Wagner. 2023. "Nanodrugs for the Treatment of Ischemic Stroke: A Systematic Review" International Journal of Molecular Sciences 24, no. 13: 10802. https://doi.org/10.3390/ijms241310802

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop