The Role of microRNAs in Neurological Pathologies: Comparison
Please note this is a comparison between Version 2 by Jessie Wu and Version 4 by Jessie Wu.

MicroRNAs (miRNAs) are small non-coding RNA molecules that are 18–25 nucleotides long (22 nucleotides on average) and involved in the transcriptional and post-transcriptional regulation of gene expression by RNA interference, which is of great interest to molecular biologists, geneticists, and biochemists. These molecules are mainly present intracellularly, but there is also an extracellular (circulating) microRNA fraction. The existence and functions of more than 2500 human miRNAs are known. 

  • miRNA
  • Alzheimer’s disease
  • ischemia–reperfusion injury
  • blood–brain barrier
  • RNA biology
  • neuroinflammation

1. The Role of MicroRNAs in Signaling Pathways of Alzheimer’s-Type Neurodegeneration

Disruption of epigenetic regulation and alterations in microRNA expression, which usually occur in conjunction, are important factors in the pathogenesis of many neurological disorders [1][2].
Several recent and extensive systematic reviews [3][4][5][6][7][8] have addressed the role of microRNAs in the pathogenesis of Alzheimer’s disease. These reviews explore numerous microRNA variants involved in the epigenetic regulation of the neurodegenerative process that serve as potential targets for diagnosis and targeted therapy.
Examples of such microRNAs include the molecules miR-200b, miR-135a, miR-10a-5p, miR-142a-5p, miR-146a-5p, miR-155-5p, miR-211-5p, miR-455-5p, miR-34a, miR-125b, miR-181c, miR-9, miR-191-5p, miR-181c, and miR-206, which are considered potential diagnostic markers for Alzheimer’s disease [9]. Additionally, attention should be paid to microRNA molecules that possess both diagnostic and therapeutic potential: miR-128, miR-574, miR-146-a, miR-181, miR-132, miR-188-5p, and miR-137 [9].
There are also studies describing modern bioinformatic approaches that use artificial intelligence and machine learning algorithms for identifying new biomarkers and improving the accuracy of molecular diagnosis of Alzheimer’s disease [10][11][12][13][14].
Some review papers present important data on other non-coding RNAs, such as circular RNAs and long non-coding RNAs, which participate in the pathogenesis of neurodegeneration and regulate the interplay between microRNAs/mRNAs/regulatory signaling pathways, which are mediated genetically [15][16]. These RNA molecules are often considered by researchers as more efficient targets for diagnostic and therapeutic strategies. Another important contemporary research direction in Alzheimer’s disease pathogenesis is the study of the regulatory functions of both microRNAs localized in specific organelles (such as mitochondria [17]) and exosomal microRNAs [18][19].
The assessment of neurovascular dysfunction is of particular interest as it plays a crucial role in the onset and progression of the neurodegenerative process associated with the accumulation of β-amyloid peptide in brain neurons and cerebral vessel walls [20][21]. Special attention is given to the dysfunction of the blood–brain barrier, reduced cerebral blood flow, and impaired vascular clearance of beta-amyloid from the brain into the glymphatic system and meningeal lymphatic vessels [22]. These disturbances may, in turn, be linked to the reprogramming of epigenetic regulation [23][24].
According to several authors, RNA interference mediated by microRNAs can initiate neurovascular events leading to Alzheimer’s-type neurodegeneration [25][26][27][28]. This provides a strong theoretical basis for the development of new directions in targeted genetic therapy for neurodegenerative diseases [29][30][31][32].

2. Regulation of Signaling Pathways in Ischemia and Reperfusion Injury of Nerve Cells Involving microRNAs

In in vitro models, it has been shown that the activation of miR-496 and miR-874-3p reduces the consequences of ischemic–reperfusion injury in neurons by negatively regulating BCL2L14 and BMF/BCL2L13, respectively [33][34]. Activation of miR-92b-3p expression inhibits apoptosis, mitochondrial dysfunction, and inflammation through the inhibition of TRAF3 [35]. MiR-182-5p and miR-193b-3p also exert neuroprotective effects in cerebral ischemia–reperfusion injury by negatively regulating the inflammatory response mediated by Toll-like receptor 4 and 5-lipoxygenase, respectively [36][37]. Additionally, miR-30c acts by inhibiting neuronal apoptosis in ischemia–reperfusion injury of the brain, thus suppressing the expression of SOX9/MAPK [38][39], while miR-449a downregulates the expression of amphiregulin (AREG) [40].
Similar effects are observed with the inhibition of miR-19a-3p, which reduces the extent and area of cerebral ischemia–reperfusion injury by regulating inflammation and apoptosis through increased expression of IGFBP3 both in vivo and in vitro [41]. It has also been discovered that inhibiting exosomal miR-200a-3p/141-3p, which originates from astrocytes and targets the gene SIRT1 and its associated signaling pathways, reduces the pathological consequences of cerebral ischemia–reperfusion injury in a mouse model [42]. MiR-370 has been shown to exacerbate neuronal reperfusion injury by impacting the expression of SIRT6 and the regulation mechanism of the Nrf2/ARE signaling pathway [43], while the exosomal form of this microRNA (370-3p) increases blood–brain barrier permeability during ischemia–reperfusion injury through the interference of MAPK1 [44].
Long non-coding RNA MEG3, by binding to miR-485, promotes the exacerbation of cerebral ischemia–reperfusion injury through potentiating pyroptosis via AIM2 [45]. Additionally, the knockdown of miR-155-5p acts by inhibiting the pyroptosis mechanism and inflammation through interference with DUSP14 [46][47]. It should be noted that Zhang L and colleagues [48], without emphasizing the orientation of miR-155, also demonstrated its activating influence on apoptosis and inflammatory processes in neural tissue.
In addition to the mentioned studies, there are several other works concerning the role of microRNAs in the pathogenesis of ischemia–reperfusion injury and the signaling pathways through which their effects are realized during RNA interference (Figure 1). The majority of neuroprotective mechanisms affected by the overexpression or inhibition of microRNAs involve anti-apoptotic, anti-inflammatory, and antioxidant signaling pathways.
Figure 1. Examples of effects of various microRNA (miRNAs)s on signaling pathways in the pathogenesis of ischemia–reperfusion brain injury. EPR = endoplasmic reticulum; BBB = blood–brain barrier; green arrows = positive biological effect of RNA interference; red arrows = negative biological effect of RNA interference. The dark blue color of the molecules indicates microRNA-3p (sense); the olive color indicates microRNA-5p (antisense); the aquamarine color indicates microRNAs with an unknown formation end from pre-microRNA.
Most studies from various researchers have demonstrated unidirectional effectiveness regarding the neuroprotective role of miR-10b-3p [49][50], miR-124-3p [51][52], miR-141-3p [42][53], miR-153-3p [54][55], miR-182-5p [36][56], miR-20a-3p [57][58], miR-22-3p [59][60], miR-24-3p [61][62], miR-27a-3p [63][64][65], miR-342-5p [66][67], miR-455-3p [68][69], miR-488-3p [70][71], and miR-92b-3p [35][72] as well as the damaging role of miR-141-3p [42][53], miR-155-5p [46][47], and miR-182 [73][74]. Therefore, these specific microRNAs should be considered as priority targets for further translation into clinical practice.
Indeed, along with the discovery of unidirectional effects of microRNA expression, contradictory results concerning the same molecules are also encountered. For example, according to the findings presented by Jia T et al. [75], activation of miR-489-3p expression in in vivo and in vitro models reduces the pathological consequences of cerebral ischemia–reperfusion by inhibiting histone deacetylase 2 (HDAC2), thereby reducing apoptosis intensity and enhancing cell viability. In contrast, Song L et al. [76] obtained opposing results: in mice subjected to temporary middle cerebral artery occlusion, an intensification of oxidative stress and neuron apoptosis was observed under conditions of increased miR-489-3p levels, which inhibits Sirtuin1 (SIRT1).
In the study investigating the effects of miR-421-3p in cerebral ischemia–reperfusion [77], both in vivo and in vitro models showed that increased expression of miR-421-3p reduces the intensity of inflammation through the YTHDF1/NF-κB p65 signaling pathway. On the contrary, Xu J. et al. [78] found a reverse effect of this microRNA concerning the intensity of apoptosis and inflammation in models of ischemia–reperfusion nerve tissue damage, mediated through the myocyte enhancer factor 2C (MEF2C).
In the study by Wei XY et al. [79], positive effects of the long non-coding RNA (lncRNA) RPL34-AS1 were observed in patients with stroke as well as in cerebral ischemia in rats and in an OGD cell model. This molecule inhibits miR-223-3p, which targets the insulin-like growth factor 1 receptor (IGF1R). The authors attribute the positive effects of RPL34-AS1 to its influence on this specific mechanism. However, there are contradictory results regarding the effects of miR-223-3p in studies focusing on the circular RNA circPDS5B and its impact on angiogenesis [80] and regarding the positive impact of miR-223-3p expression on the development of the inflammatory response in the zone of ischemia–reperfusion and its surrounding area [81].
The evaluation of the effects of miR-19a and its sense form miR-19a-3p shows a negative role in the mechanism of cerebral ischemia–reperfusion injury played by excessive stimulation of apoptosis and inflammation [41][82][83]. Similar observations are confirmed for the structurally related miR-19b-3p, which also intensifies the neuroinflammatory process in the ischemia–reperfusion zone [83]. However, these observations are contradicted by data showing that knockdown of the long non-coding RNA H19 and overexpression of miR-19a-3p attenuated the severity of ischemia–reperfusion-induced oxidative stress and apoptosis in neurons, as reported by Gao N. et al. [84].
The study of the effects of the lncRNA Malat1 revealed a positive influence of miR-26b [85] and a negative impact of miR-142-3p expression [86] on apoptosis and cell proliferation during experimental brain hypoxia–ischemia. However, the results from Li J. et al. [87] show an opposite effect when miR-142-3p expression is enhanced: inhibiting FBXO3. It is important to note that this researchtudy has limitations as it was conducted only on an in vitro model.
In two studies investigating the role of miR-140-3p on in vitro models with OGD, opposite effects were demonstrated: on the PC12 cell line, an enhancement of miR-140-3p expression showed a weakening effect on apoptosis and oxidative stress [88], while on the N2a cell line, an overexpression of miR-140-3p potentiated apoptosis and oxidative stress [89]. Supporting the greater significance of the first study and the neuroprotective role of this molecule is the fact that the in vitro results obtained by Zhang Y. et al. were replicated in the study on patients with ischemic stroke.
Another contradiction in determining the role of miRNAs in the pathogenesis of ischemia–reperfusion brain injury is the data on miR-128-3p. They indicate its proapoptotic efficacy in in vitro and in vivo models by inhibiting the FOXO/Relaxin signaling pathway [90] as well as the neuroprotective efficacy (potentiation of differentiation and myelination processes) of this miRNA in exosomal localization in an in vivo experiment [91].
Moreover, the studies mentioned above demonstrate that miR-135a, miR-181c, and miR-211-5p, whose expression plays a positive role in protection against cerebral ischemia–reperfusion injury, also act as neuroprotective agents in Alzheimer’s disease [9]. Conversely, the overexpression of miR-155-5p, miR-200a-3p, and miR-206 leads to damage to neural tissue both in the context of ischemia–reperfusion and Alzheimer’s-like neurodegeneration [9].

References

  1. Neal, M.; Richardson, J.R. Epigenetic regulation of astrocyte function in neuroinflammation and neurodegeneration. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 432–443.
  2. Ghosh, P.; Saadat, A. Neurodegeneration and epigenetics: A review. Neurologia 2023, 38, e62–e68.
  3. Takousis, P.; Sadlon, A.; Schulz, J.; Wohlers, I.; Dobricic, V.; Middleton, L.; Lill, C.M.; Perneczky, R.; Bertram, L. Differential expression of microRNAs in Alzheimer’s disease brain, blood, and cerebrospinal fluid. Alzheimer’s Dement. 2019, 15, 1468–1477.
  4. Swarbrick, S.; Wragg, N.; Ghosh, S.; Stolzing, A. Systematic Review of miRNA as Biomarkers in Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 6156–6167.
  5. Nunomura, A.; Perry, G. RNA and Oxidative Stress in Alzheimer’s Disease: Focus on microRNAs. Oxidative Med. Cell. Longev. 2020, 2020, 2638130.
  6. Walgrave, H.; Zhou, L.; De Strooper, B.; Salta, E. The promise of microRNA-based therapies in Alzheimer’s disease: Challenges and perspectives. Mol. Neurodegener. 2021, 16, 76.
  7. Mayo, S.; Benito-León, J.; Peña-Bautista, C.; Baquero, M.; Cháfer-Pericás, C. Recent Evidence in Epigenomics and Proteomics Biomarkers for Early and Minimally Invasive Diagnosis of Alzheimer’s and Parkinson’s Diseases. Curr. Neuropharmacol. 2021, 19, 1273–1303.
  8. Liu, S.; Fan, M.; Zheng, Q.; Hao, S.; Yang, L.; Xia, Q.; Qi, C.; Ge, J. MicroRNAs in Alzheimer’s disease: Potential diagnostic markers and therapeutic targets. Biomed. Pharmacother. 2022, 148, 112681.
  9. Silvestro, S.; Bramanti, P.; Mazzon, E. Role of miRNAs in Alzheimer’s Disease and Possible Fields of Application. Int. J. Mol. Sci. 2019, 20, 3979.
  10. Yuen, S.C.; Liang, X.; Zhu, H.; Jia, Y.; Leung, S.W. Prediction of differentially expressed microRNAs in blood as potential biomarkers for Alzheimer’s disease by meta-analysis and adaptive boosting ensemble learning. Alzheimer’s Res. Ther. 2021, 13, 126.
  11. Chang, C.H.; Lin, C.H.; Lane, H.Y. Machine Learning and Novel Biomarkers for the Diagnosis of Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 2761.
  12. Xu, A.; Kouznetsova, V.L.; Tsigelny, I.F. Alzheimer’s Disease Diagnostics Using MiRNA Biomarkers and Machine Learning. J. Alzheimer’s Dis. 2022, 86, 841–859.
  13. Shokhirev, M.N.; Johnson, A.A. An integrative machine-learning meta-analysis of high-throughput omics data identifies age-specific hallmarks of Alzheimer’s disease. Ageing Res. Rev. 2022, 81, 101721.
  14. Chiricosta, L.; D’Angiolini, S.; Gugliandolo, A.; Mazzon, E. Artificial Intelligence Predictor for Alzheimer’s Disease Trained on Blood Transcriptome: The Role of Oxidative Stress. Int. J. Mol. Sci. 2022, 23, 5237.
  15. Akhter, R. Circular RNA and Alzheimer’s Disease. Adv. Exp. Med. Biol. 2018, 1087, 239–243.
  16. Ma, N.; Tie, C.; Yu, B.; Zhang, W.; Wan, J. Identifying lncRNA-miRNA-mRNA networks to investigate Alzheimer’s disease pathogenesis and therapy strategy. Aging 2020, 12, 2897–2920.
  17. Gowda, P.; Reddy, P.H.; Kumar, S. Deregulated mitochondrial microRNAs in Alzheimer’s disease: Focus on synapse and mitochondria. Ageing Res. Rev. 2022, 73, 101529.
  18. Su, L.; Li, R.; Zhang, Z.; Liu, J.; Du, J.; Wei, H. Identification of altered exosomal microRNAs and mRNAs in Alzheimer’s disease. Ageing Res. Rev. 2022, 73, 101497.
  19. Garcia, G.; Pinto, S.; Ferreira, S.; Lopes, D.; Serrador, M.J.; Fernandes, A.; Vaz, A.R.; Mendonça, A.; Edenhofer, F.; Malm, T.; et al. Emerging Role of miR-21-5p in Neuron-Glia Dysregulation and Exosome Transfer Using Multiple Models of Alzheimer’s Disease. Cells 2022, 11, 3377.
  20. Ryazanova, M.V.; Averchuk, A.S.; Novikova, S.V.; Salmina, A.B. Molecular mechanisms of angiogenesis: Brain is in the focus. Opera Med. Physiol. 2022, 9, 54–72.
  21. Tregub, P.P.; Averchuk, A.S.; Baranich, T.I.; Ryazanova, M.V.; Salmina, A.B. Physiological and Pathological Remodeling of Cerebral Microvessels. Int. J. Mol. Sci. 2022, 23, 12683.
  22. Da Mesquita, S.; Louveau, A.; Vaccari, A.; Smirnov, I.; Cornelison, R.C.; Kingsmore, K.M.; Contarino, C.; Onengut-Gumuscu, S.; Farber, E.; Raper, D.; et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 2018, 560, 185–191.
  23. Ihezie, S.A.; Mathew, I.E.; McBride, D.W.; Dienel, A.; Blackburn, S.L.; Thankamani Pandit, P.K. Epigenetics in blood-brain barrier disruption. Fluids Barriers CNS 2021, 18, 17.
  24. Neag, M.A.; Mitre, A.O.; Burlacu, C.C.; Inceu, A.I.; Mihu, C.; Melincovici, C.S.; Bichescu, M.; Buzoianu, A.D. miRNA Involvement in Cerebral Ischemia-Reperfusion Injury. Front. Neurosci. 2022, 16, 901360.
  25. Van den Hove, D.L.; Kompotis, K.; Lardenoije, R.; Kenis, G.; Mill, J.; Steinbusch, H.W.; Lesch, K.P.; Fitzsimons, C.P.; De Strooper, B.; Rutten, B.P. Epigenetically regulated microRNAs in Alzheimer’s disease. Neurobiol. Aging 2014, 35, 731–745.
  26. Stoccoro, A.; Coppedè, F. Role of epigenetics in Alzheimer’s disease pathogenesis. Neurodegener. Dis. Manag. 2018, 8, 181–193.
  27. Nikolac Perkovic, M.; Videtic Paska, A.; Konjevod, M.; Kouter, K.; Svob Strac, D.; Nedic Erjavec, G.; Pivac, N. Epigenetics of Alzheimer’s Disease. Biomolecules 2021, 11, 195.
  28. Bryzgalov, L.O.; Korbolina, E.E.; Merkulova, T.I. Exploring the Genetic Predisposition to Epigenetic Changes in Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 7955.
  29. Coneys, R.; Wood, I.C. Alzheimer’s disease: The potential of epigenetic treatments and current clinical candidates. Neurodegener. Dis. Manag. 2020, 10, 543–558.
  30. Pandey, D.; Pal, T.; Sharma, A.; Flora, S. Potential Epigenetic Targets for Combating Alzheimer’s Disease. Mini Rev. Med. Chem. 2021, 21, 1527–1540.
  31. Coppedè, F. Epigenetic regulation in Alzheimer’s disease: Is it a potential therapeutic target? Expert Opin. Ther. Targets 2021, 25, 283–298.
  32. Hajjo, R.; Sabbah, D.A.; Abusara, O.H.; Al Bawab, A.Q. A Review of the Recent Advances in Alzheimer’s Disease Research and the Utilization of Network Biology Approaches for Prioritizing Diagnostics and Therapeutics. Diagnostics 2022, 12, 2975.
  33. Yao, X.; Yao, R.; Yi, J.; Huang, F. Upregulation of miR-496 decreases cerebral ischemia/reperfusion injury by negatively regulating BCL2L14. Neurosci. Lett. 2019, 696, 197–205.
  34. Jiang, D.; Sun, X.; Wang, S.; Man, H. Upregulation of miR-874-3p decreases cerebral ischemia/reperfusion injury by directly targeting BMF and BCL2L13. Biomed. Pharmacother. 2019, 117, 108941.
  35. Liu, E.; Sun, H.; Wu, J.; Kuang, Y. MiR-92b-3p regulates oxygen and glucose deprivation-reperfusion-mediated apoptosis and inflammation by targeting TRAF3 in PC12 cells. Exp. Physiol. 2020, 105, 1792–1801.
  36. Wang, J.; Xu, Z.; Chen, X.; Li, Y.; Chen, C.; Wang, C.; Zhu, J.; Wang, Z.; Chen, W.; Xiao, Z.; et al. MicroRNA-182-5p attenuates cerebral ischemia-reperfusion injury by targeting Toll-like receptor 4. Biochem. Biophys. Res. Commun. 2018, 505, 677–684.
  37. Chen, Z.; Yang, J.; Zhong, J.; Luo, Y.; Du, W.; Hu, C.; Xia, H.; Li, Y.; Zhang, J.; Li, M.; et al. MicroRNA-193b-3p alleviates focal cerebral ischemia and reperfusion-induced injury in rats by inhibiting 5-lipoxygenase expression. Exp. Neurol. 2020, 327, 113223.
  38. Zhang, M.; Zhu, Y.; Wei, M.; Liu, H. Neuroprotective effects of miR-30c on rats with cerebral ischemia/reperfusion injury by targeting SOX9. Pathol. Res. Pract. 2020, 216, 153271.
  39. Zhang, H.; Li, M.; Liang, J.; Li, M.; Sun, X. Long Non-coding RNA PVT1 Inhibits miR-30c-5p to Upregulate Rock2 to Modulate Cerebral Ischemia/Reperfusion Injury Through MAPK Signaling Pathway Activation. Mol. Neurobiol. 2021, 58, 6032–6048.
  40. Yu, Y.; Zhang, X.; Han, Z.; Zhao, W.; Zhang, L. Expression and regulation of miR-449a and AREG in cerebral ischemic injury. Metab. Brain Dis. 2019, 34, 821–832.
  41. Chai, Z.; Gong, J.; Zheng, P.; Zheng, J. Inhibition of miR-19a-3p decreases cerebral ischemia/reperfusion injury by targeting IGFBP3 in vivo and in vitro. Biol. Res. 2020, 53, 17.
  42. Wei, W.; Li, H.; Deng, Y.; Zheng, X.; Zhou, Y.; Xue, X. The combination of Alisma and Atractylodes ameliorates cerebral ischaemia/reperfusion injury by negatively regulating astrocyte-derived exosomal miR-200a-3p/141-3p by targeting SIRT1. J. Ethnopharmacol. 2023, 313, 116597.
  43. Ruan, Z.F.; Xie, M.; Gui, S.J.; Lan, F.; Wan, J.; Li, Y. MiR-370 accelerated cerebral ischemia reperfusion injury via targeting SIRT6 and regulating Nrf2/ARE signal pathway. Kaohsiung J. Med. Sci. 2020, 36, 741–749.
  44. Gu, C.; Mo, W.; Wang, K.; Gao, M.; Chen, J.; Zhang, F.; Shen, J. Exosomal miR-370-3p increases the permeability of blood-brain barrier in ischemia/reperfusion stroke of brain by targeting MPK1. Aging 2023, 15, 1931–1943.
  45. Liang, J.; Wang, Q.; Li, J.Q.; Guo, T.; Yu, D. Long non-coding RNA MEG3 promotes cerebral ischemia-reperfusion injury through increasing pyroptosis by targeting miR-485/AIM2 axis. Exp. Neurol. 2020, 325, 113139.
  46. Shi, Y.; Li, K.; Xu, K.; Liu, Q.H. MiR-155-5p accelerates cerebral ischemia-reperfusion injury via targeting DUSP14 by regulating NF-κB and MAPKs signaling pathways. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1408–1419.
  47. Shi, Y.; Li, Z.; Li, K.; Xu, K. miR-155-5p accelerates cerebral ischemia-reperfusion inflammation injury and cell pyroptosis via DUSP14/ TXNIP/NLRP3 pathway. Acta Biochim. Pol. 2022, 69, 787–793.
  48. Zhang, L.; Liu, C.; Huang, C.; Xu, X.; Teng, J. miR-155 Knockdown Protects against Cerebral Ischemia and Reperfusion Injury by Targeting MafB. BioMed Res. Int. 2020, 2020, 6458204.
  49. Ye, X.; Fang, H.; Feng, Y.; Qian, H.; Wu, Z.; Chen, S. MiR-10b-3p Protects Cerebral I/R Injury through Targeting Programmed Cell Death 5 (PDCD5). Crit. Rev. Eukaryot. Gene Expr. 2021, 31, 85–98.
  50. Sun, K.; Zhang, J.; Yang, Q.; Zhu, J.; Zhang, X.; Wu, K.; Li, Z.; Xie, W.; Luo, X. MiR-10b-3p alleviates cerebral ischemia/reperfusion injury by targeting Krüppel-like factor 5 (KLF5). Pflügers Arch. -Eur. J. Physiol. 2022, 474, 343–353.
  51. Huang, P.; Wei, S.; Ren, J.; Tang, Z.; Guo, M.; Situ, F.; Zhang, D.; Zhu, J.; Xiao, L.; Xu, J.; et al. MicroRNA-124-3p alleviates cerebral ischaemia-induced neuroaxonal damage by enhancing Nrep expression. J. Stroke Cerebrovasc. Dis. 2023, 32, 106949.
  52. Zhang, K.L.; Li, S.M.; Hou, J.Y.; Hong, Y.H.; Chen, X.X.; Zhou, C.Q.; Wu, H.; Zheng, G.H.; Zeng, C.T.; Wu, H.D.; et al. Elabela, a Novel Peptide, Exerts Neuroprotective Effects Against Ischemic Stroke Through the APJ/miR-124-3p/CTDSP1/AKT Pathway. Cell. Mol. Neurobiol. 2023, 43, 2989–3003.
  53. Liu, T.; Feng, J.; Sun, Z.; He, M.; Sun, L.; Dong, S.; Guo, Z.; Zhang, G. Inhibition of miR-141-3p attenuates apoptosis of neural stem cells via targeting PBX1 to regulate PROK2 transcription in MCAO mice. Cell Cycle 2023, 22, 403–418.
  54. Li, Y.; Peng, B.; Li, Y.; Huang, A.; Peng, Y.; Yu, Q.; Li, Y. MiR-203a-3p/153-3p improves cognitive impairments induced by ischemia/reperfusion via blockade of SRC-mediated MAPK signaling pathway in ischemic stroke. Chem. Biol. Interact. 2022, 358, 109900.
  55. Wang, H.J.; Tang, X.L.; Huang, G.; Li, Y.B.; Pan, R.H.; Zhan, J.; Wu, Y.K.; Liang, J.F.; Bai, X.X.; Cai, J. Long Non-Coding KCNQ1OT1 Promotes Oxygen-Glucose-Deprivation/Reoxygenation-Induced Neurons Injury Through Regulating MIR-153-3p/FOXO3 Axis. J. Stroke Cerebrovasc. Dis. 2020, 29, 105126.
  56. Ding, W.; Gu, Q.; Liu, M.; Zou, J.; Sun, J.; Zhu, J. Astrocytes-derived exosomes pre-treated by berberine inhibit neuroinflammation after stroke via miR-182-5p/Rac1 pathway. Int. Immunopharmacol. 2023, 118, 110047.
  57. Sampath, D.; Branyan, T.E.; Markowsky, K.G.; Gunda, R.; Samiya, N.; Obenaus, A.; Sohrabji, F. Sex differences in cognitive impairment after focal ischemia in middle-aged rats and the effect of iv miR-20a-3p treatment. Neurobiol. Aging 2023, 129, 168–177.
  58. Branyan, T.E.; Selvamani, A.; Park, M.J.; Korula, K.E.; Kosel, K.F.; Srinivasan, R.; Sohrabji, F. Functional Assessment of Stroke-Induced Regulation of miR-20a-3p and Its Role as a Neuroprotectant. Transl. Stroke Res. 2022, 13, 432–448.
  59. Zhang, Y.; Liu, J.; Su, M.; Wang, X.; Xie, C. Exosomal microRNA-22-3p alleviates cerebral ischemic injury by modulating KDM6B/BMP2/BMF axis. Stem Cell Res. Ther. 2021, 12, 111.
  60. Zhang, H.S.; Ouyang, B.; Ji, X.Y.; Liu, M.F. Gastrodin Alleviates Cerebral Ischaemia/Reperfusion Injury by Inhibiting Pyroptosis by Regulating the lncRNA NEAT1/miR-22-3p Axis. Neurochem. Res. 2021, 46, 1747–1758.
  61. Kuai, F.; Zhou, L.; Zhou, J.; Sun, X.; Dong, W. Long non-coding RNA THRIL inhibits miRNA-24-3p to upregulate neuropilin-1 to aggravate cerebral ischemia-reperfusion injury through regulating the nuclear factor κB p65 signaling. Aging 2021, 13, 9071–9084.
  62. Di, G.; Yang, X.; Cheng, F.; Liu, H.; Xu, M. CEBPA-AS1 Knockdown Alleviates Oxygen-Glucose Deprivation/Reperfusion-Induced Neuron Cell Damage by the MicroRNA 24-3p/BOK Axis. Mol. Cell. Biol. 2021, 41, e0006521.
  63. Li, W.; Zhu, Q.; Xu, X.; Hu, X. MiR-27a-3p suppresses cerebral ischemia-reperfusion injury by targeting FOXO1. Aging 2021, 13, 11727–11737.
  64. Zhang, Z.; He, J.; Wang, B. Circular RNA circ_HECTD1 regulates cell injury after cerebral infarction by miR-27a-3p/FSTL1 axis. Cell Cycle 2021, 20, 914–926.
  65. Li, J.; Peng, L.; Bai, W.; Peng, P.; Chen, W.; Yang, W.; Shao, J. Biliverdin Protects Against Cerebral Ischemia/Reperfusion Injury by Regulating the miR-27a-3p/Rgs1 Axis. Neuropsychiatr. Dis Treat. 2021, 17, 1165–1181.
  66. Yu, Z.; Zhu, M.; Shu, D.; Zhang, R.; Xiang, Z.; Jiang, A.; Liu, S.; Zhang, C.; Yuan, Q.; Hu, X. LncRNA PEG11as aggravates cerebral ischemia/reperfusion injury after ischemic stroke through miR-342-5p/PFN1 axis. Life Sci. 2023, 313, 121276.
  67. Zhu, H.; Zhang, Y.; Zhu, Y. MiR-342-5p protects neurons from cerebral ischemia induced-apoptosis through regulation of Akt/NF-κB pathways by targeting CCAR2. J. Stroke Cerebrovasc. Dis. 2023, 32, 106901.
  68. Gan, C.; Ouyang, F. Exosomes Released from Bone-Marrow Stem Cells Ameliorate Hippocampal Neuronal Injury Through transferring miR-455-3p. J. Stroke Cerebrovasc. Dis. 2022, 31, 106142.
  69. Fan, Y.; Wei, L.; Zhang, S.; Song, X.; Yang, J.; He, X.; Zheng, X. LncRNA SNHG15 Knockdown Protects Against OGD/R-Induced Neuron Injury by Downregulating TP53INP1 Expression via Binding to miR-455-3p. Neurochem. Res. 2021, 46, 1019–1030.
  70. Zheng, H.; Zhang, G.; Liu, G.; Wang, L. Up-regulation of lncRNA NEAT1 in cerebral ischemic stroke promotes activation of astrocytes by modulation of miR-488-3p/RAC1. Exp. Brain Res. 2023, 241, 395–406.
  71. Zhou, L.; Yang, W.; Yao, E.; Li, H.; Wang, J.; Wang, K.; Zhong, X.; Peng, Z.; Huang, X. MicroRNA-488-3p Regulates Neuronal Cell Death in Cerebral Ischemic Stroke Through Vacuolar Protein Sorting 4B (VPS4B). Neuropsychiatr. Dis. Treat. 2021, 17, 41–55.
  72. Huang, Y.; Tang, J.; Li, X.; Long, X.; Huang, Y.; Zhang, X. miR-92b-3p Exerts Neuroprotective Effects on Ischemia/Reperfusion-Induced Cerebral Injury via Targeting NOX4 in a Rat Model. Oxidative Med. Cell. Longev. 2022, 2022, 3494262.
  73. Zhang, T.; Tian, C.; Wu, J.; Zhang, Y.; Wang, J.; Kong, Q.; Mu, L.; Sun, B.; Ai, T.; Wang, Y.; et al. MicroRNA-182 exacerbates blood-brain barrier (BBB) disruption by downregulating the mTOR/FOXO1 pathway in cerebral ischemia. FASEB J. 2020, 34, 13762–13775.
  74. Alhadidi, Q.M.; Xu, L.; Sun, X.; Althobaiti, Y.S.; Almalki, A.; Alsaab, H.O.; Stary, C.M. MiR-182 Inhibition Protects Against Experimental Stroke in vivo and Mitigates Astrocyte Injury and Inflammation in vitro via Modulation of Cortactin Activity. Neurochem. Res. 2022, 47, 3682–3696.
  75. Jia, T.; Wang, M.; Yan, W.; Wu, W.; Shen, R. Upregulation of miR-489-3p Attenuates Cerebral Ischemia/Reperfusion Injury by Targeting Histone Deacetylase 2 (HDAC2). Neuroscience 2022, 484, 16–25.
  76. Song, L.; Mu, L.; Wang, H. MicroRNA-489-3p aggravates neuronal apoptosis and oxidative stress after cerebral ischemia-reperfusion injury. Bioengineered 2022, 13, 14047–14056.
  77. Zheng, L.; Tang, X.; Lu, M.; Sun, S.; Xie, S.; Cai, J.; Zan, J. microRNA-421-3p prevents inflammatory response in cerebral ischemia/reperfusion injury through targeting m6A Reader YTHDF1 to inhibit p65 mRNA translation. Int. Immunopharmacol. 2020, 88, 106937.
  78. Xu, J.; Huang, X.; Liu, S.; Chen, D.; Xie, Y.; Zhao, Z. The protective effects of lncRNA ZFAS1/miR-421-3p/MEF2C axis on cerebral ischemia-reperfusion injury. Cell Cycle 2022, 21, 1915–1931.
  79. Wei, X.Y.; Zhang, T.Q.; Suo, R.; Qu, Y.Y.; Chen, Y.; Zhu, Y.L. Long non-coding RNA RPL34-AS1 ameliorates oxygen-glucose deprivation-induced neuronal injury via modulating miR-223-3p/IGF1R axis. Hum. Cell 2022, 35, 1785–1796.
  80. Kui, L.; Li, Z.; Wang, G.; Li, X.; Zhao, F.; Jiao, Y. CircPDS5B Reduction Improves Angiogenesis Following Ischemic Stroke by Regulating MicroRNA-223-3p/NOTCH2 Axis. Neurol. Genet. 2023, 9, e200074.
  81. Zhao, Y.; Gan, Y.; Xu, G.; Hua, K.; Liu, D. Exosomes from MSCs overexpressing microRNA-223-3p attenuate cerebral ischemia through inhibiting microglial M1 polarization mediated inflammation. Life Sci. 2020, 260, 118403.
  82. Ge, X.L.; Wang, J.L.; Liu, X.; Zhang, J.; Liu, C.; Guo, L. Inhibition of miR-19a protects neurons against ischemic stroke through modulating glucose metabolism and neuronal apoptosis. Cell. Mol. Biol. Lett. 2019, 24, 37.
  83. Zhou, F.; Wang, Y.K.; Zhang, C.G.; Wu, B.Y. miR-19a/b-3p promotes inflammation during cerebral ischemia/reperfusion injury via SIRT1/FoxO3/SPHK1 pathway. J. Neuroinflamm. 2021, 18, 122.
  84. Gao, N.; Tang, H.; Gao, L.; Tu, G.L.; Luo, H.; Xia, Y. LncRNA H19 Aggravates Cerebral Ischemia/Reperfusion Injury by Functioning as a ceRNA for miR-19a-3p to Target PTEN. Neuroscience 2020, 437, 117–129.
  85. Li, Z.; Li, J.; Tang, N. Long noncoding RNA Malat1 is a potent autophagy inducer protecting brain microvascular endothelial cells against oxygen-glucose deprivation/reoxygenation-induced injury by sponging miR-26b and upregulating ULK2 expression. Neuroscience 2017, 354, 1–10.
  86. Meng, S.; Wang, B.; Li, W. LncRNA MALAT1 improves cerebral ischemia-reperfusion injury and cognitive dysfunction by regulating miR-142-3p/SIRT1 axis. Int. J. Neurosci. 2023, 133, 740–753.
  87. Li, J.; Ma, L. MiR-142-3p Attenuates Oxygen Glucose Deprivation/Reoxygenation-Induced Injury by Targeting FBXO3 in Human Neuroblastoma SH-SY5Y Cells. World Neurosurg. 2020, 136, e149–e157.
  88. Zhang, Y.; Su, Q.; Xia, W.; Jia, K.; Meng, D.; Wang, X.; Ni, X.; Su, Z. MiR-140–3p directly targets Tyro3 to regulate OGD/R-induced neuronal injury through the PI3K/Akt pathway. Brain Res. Bull. 2023, 192, 93–106.
  89. Yi, M.; Li, Y.; Wang, D.; Zhang, Q.; Yang, L.; Yang, C. KCNQ1OT1 Exacerbates Ischemia-Reperfusion Injury Through Targeted Inhibition of miR-140-3P. Inflammation 2020, 43, 1832–1845.
  90. Yan, Q.; Sun, S.Y.; Yuan, S.; Wang, X.Q.; Zhang, Z.C. Inhibition of microRNA-9-5p and microRNA-128-3p can inhibit ischemic stroke-related cell death in vitro and in vivo. IUBMB Life 2020, 72, 2382–2390.
  91. Hou, H.; Wang, Y.; Yang, L.; Wang, Y. Exosomal miR-128-3p reversed fibrinogen-mediated inhibition of oligodendrocyte progenitor cell differentiation and remyelination after cerebral ischemia. CNS Neurosci. Ther. 2023, 29, 1405–1422.
More
Video Production Service