1
|
An Y, Su G, Chen W, Song J, Chai M, Zhu L, Zhang Z. Research progress on the mechanisms of microglial extracellular vesicles affecting the prognosis of ischemic stroke. Neurochem Int 2025; 185:105949. [PMID: 40015338 DOI: 10.1016/j.neuint.2025.105949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/26/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
Ischemic stroke is the major type of stroke and one of the main causes of morbidity, mortality, and long-term disability worldwide. Microglia play a complex and crucial role in stroke. They are the primary immune cells in the brain and can rapidly respond to the pathological changes caused by stroke. They promote neuroprotection and repair after ischemic stroke through various mechanisms, such as activation and polarization, dynamic interactions with other cells (neurons, astrocytes, oligodendrocytes, vascular endothelial cells, etc.), and phagocytosis to clear dead cell debris. Among the multiple pathways through which microglia exert their neuroprotective effects, the secretion of extracellular vesicles is one of the most important. The focus of this review is to analyze the latest progress in research on ischemic stroke related to microglia-derived extracellular vesicles, discuss their mechanisms of action, and provide new strategies for improving stroke prognosis. To obtain relevant articles, we conducted a comprehensive search in Pubmed and Web of Science, with keywords related to ischemic stroke and microglia-derived extracellular vesicles or exosomes. A total of 59 articles were included in the review. Existing studies have shown that after a stroke occurs, microglia release extracellular vesicles containing proteins, nucleic acids, metabolites, etc. These vesicles target corresponding receptor cells and can slow down the development of stroke and improve stroke outcomes through various means, such as reducing neuronal apoptosis, inhibiting neuronal autophagy, suppressing neuronal ferroptosis, preventing neuronal pyroptosis, alleviating inflammatory responses, reducing glial scar formation, promoting myelin regeneration and repair, and facilitating blood-brain barrier repair.
Collapse
Affiliation(s)
- Yang An
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, Gansu, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, 730030 Lanzhou, Gansu, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, Gansu, China
| | - Jinyang Song
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, Gansu, China
| | - Miao Chai
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, Gansu, China
| | - Longni Zhu
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, Gansu, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Mathias K, Machado RS, Petronilho T, Sulzbacher VAR, de Rezende VL, Prophiro JS, Petronilho F. Glial and blood-brain barrier cell-derived exosomes: Implications in stroke. Microvasc Res 2025; 160:104812. [PMID: 40246225 DOI: 10.1016/j.mvr.2025.104812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Exosomes are small extracellular vesicles released by cells that play a pivotal role in intercellular communication, significantly influencing both the pathophysiology and potential treatment of ischemic stroke (IS). This review examines exosomes derived from key brain cell types, including microglia, astrocytes, oligodendrocytes, oligodendrocyte precursor cells (NG2+ cells), endothelial cells, and pericytes, emphasizing their molecular cargo and functional impact in IS. Microglia-derived exosomes regulate neuroinflammation, with M2-type exosomes exhibiting neuroprotective effects, while astrocyte-derived exosomes modulate pathways involved in pyroptosis and autophagy, influencing neuronal survival. Oligodendrocyte and NG2+ cell-derived exosomes contribute to remyelination, axonal growth, and inflammatory modulation. Endothelial and pericyte-derived exosomes play critical roles in BBB integrity, neurovascular remodeling, and drug transport across the BBB. This synthesis highlights recent advances in understanding how exosome-mediated communication impacts IS recovery and explores their translational potential for biomarker development and targeted therapies. By manipulating exosomal composition and delivery mechanisms, novel therapeutic strategies may emerge, offering hope for improved IS treatment outcomes.
Collapse
Affiliation(s)
- Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Extremo Sul Catarinense, Criciuma, SC, Brazil; Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Extremo Sul Catarinense, Criciuma, SC, Brazil
| | - Taise Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Extremo Sul Catarinense, Criciuma, SC, Brazil
| | - Victor Augusto Rodrigues Sulzbacher
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Extremo Sul Catarinense, Criciuma, SC, Brazil
| | - Victoria Linden de Rezende
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Extremo Sul Catarinense, Criciuma, SC, Brazil
| | - Josiane Somariva Prophiro
- Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Extremo Sul Catarinense, Criciuma, SC, Brazil.
| |
Collapse
|
3
|
Gong J, Li J, Li J, He A, Ren B, Zhao M, Li K, Zhang Y, He M, Liu Y, Wang Z. Impact of Microglia-Derived Extracellular Vesicles on Resident Central Nervous System Cell Populations After Acute Brain Injury Under Various External Stimuli Conditions. Mol Neurobiol 2025:10.1007/s12035-025-04858-w. [PMID: 40126599 DOI: 10.1007/s12035-025-04858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Acute brain injuries (ABI) caused by various emergencies can lead to structural and functional damage to brain tissue. Common causes include traumatic brain injury, cerebral hemorrhage, ischemic stroke, and heat stroke. Globally, ABI represent a significant portion of neurosurgical cases. Previous studies have emphasized the significant therapeutic potential of stem cell-derived extracellular vesicles (EVs). Recent research indicates that EVs extracted from resident cells in the central nervous system (CNS) also show therapeutic potential following brain injury. Microglia, as innate immune cells of the CNS, respond to changes in the internal environment by altering their phenotype and secreting EVs that impact various CNS cells, including neurons, astrocytes, oligodendrocytes, endothelial cells, neural stem cells (NSCs), and microglia themselves. Notably, under different external stimuli, microglia can either promote neuronal survival, angiogenesis, and myelin regeneration while reducing glial scarring and inflammation, or they can exert opposite effects. This review summarizes and evaluates the current research findings on how microglia-derived EVs influence various CNS cells after ABI under different external stimuli. It analyzes the interaction mechanisms between EVs and resident CNS cells and discusses potential future research directions and clinical applications.
Collapse
Affiliation(s)
- Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Anqi He
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingcheng Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Kexin Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuchi Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mengyao He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| |
Collapse
|
4
|
Yang Q, Chen Q, Zhang KB, Liu Y, Zheng JC, Hu DX, Luo J. Sinomenine alleviates neuroinflammation in chronic cerebral hypoperfusion by promoting M2 microglial polarization and inhibiting neuronal pyroptosis via exosomal miRNA-223-3p. Acta Neuropathol Commun 2025; 13:48. [PMID: 40045356 PMCID: PMC11881310 DOI: 10.1186/s40478-025-01950-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a major contributor to vascular dementia, with neuroinflammation playing a central role in its pathogenesis. Sinomenine (SINO), a natural alkaloid derived from traditional Chinese medicine, has shown significant anti-inflammatory and neuroprotective properties. However, its efficacy and mechanism of action in CCH remain unclear. In this study, we established a CCH rat model through bilateral common carotid artery occlusion and administered 10 mg/kg of SINO daily. Behavioral tests demonstrated that SINO significantly improved cognitive and memory functions in CCH rats. Histological analysis revealed that SINO effectively reduced neuroinflammation and damage in the hippocampal CA1, CA3, and DG regions. Mechanistically, SINO promoted microglial polarization from the M1 to M2 phenotype, markedly inhibiting the release of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Further exploration of its neuroprotective mechanism showed that exosomes from SINO-treated microglia were enriched with miRNA-223-3p, which suppressed NLRP3-mediated pyroptosis in neurons. While our findings highlight the therapeutic potential of SINO, further studies are needed to validate its safety and efficacy in diverse populations and chronic settings. In summary, this study not only demonstrates SINO's regulatory effect on microglial polarization in CCH but also unveils a novel neuroprotective mechanism through exosomal miRNA-223-3p delivery, providing a solid theoretical foundation for SINO's potential as a treatment for CCH.
Collapse
Affiliation(s)
- Qu Yang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Province Key Laboratory of Precision Cell Therapy, Nanchang, Jiangxi, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qi Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Kai-Bing Zhang
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yu Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jia-Cheng Zheng
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Jiangxi Province Key Laboratory of Precision Cell Therapy, Nanchang, Jiangxi, 330006, China.
- The National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
5
|
Yang HB, Lu DC, Shu M, Li J, Ma Z. The roles and therapeutic potential of exosomal non-coding RNAs in microglia-mediated intercellular communication. Int Immunopharmacol 2025; 148:114049. [PMID: 39823800 DOI: 10.1016/j.intimp.2025.114049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/20/2025]
Abstract
Exosomes, which are small extracellular vesicles (sEVs), serve as versatile regulators of intercellular communication in the progression of various diseases, including neurological disorders. Among the diverse array of cargo they carry, non-coding RNAs (ncRNAs) play key regulatory roles in various pathophysiological processes. Exosomal ncRNAs derived from distinct cells modulate their reciprocal crosstalk locally or remotely, thereby mediating neurological diseases. Nevertheless, the emerging role of exosomal ncRNAsin microglia-mediated phenotypes remains largely unexplored. This review aims to summarise the biological functions of exosomal ncRNAs and the molecular mechanisms that underlie their impact on microglia-mediated intercellular communication, modulating neuroinflammation and synaptic functions within the landscape of neurological disorders. Furthermore, this review comprehensively described the potential applications of exosomal ncRNAs as diagnostic and prognostic biomarkers, as well as innovative therapeutic targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Hu-Bo Yang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ding-Ci Lu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Min Shu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Juan Li
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China.
| |
Collapse
|
6
|
Zhu Z, Zhang X, Lin X, Wang Y, Han C, Wang S. Research Advances and Application Progress on miRNAs in Exosomes Derived From M2 Macrophage for Tissue Injury Repairing. Int J Nanomedicine 2025; 20:1543-1560. [PMID: 39925680 PMCID: PMC11806736 DOI: 10.2147/ijn.s508781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Tissue injury repair is a multifaceted and dynamic process characterized by complex interactions among various immune cells, with M2 macrophages assuming a crucial role. Exosomes derived from M2-type macrophages (M2-Exos) significantly influence the injury repair process through intercellular communication mediated by enriched microRNAs (miRNAs). This review aims to elucidate the biological processes underlying exosome formation, the synthesis and function of miRNAs, and the diverse methodologies employed for exosome extraction. Furthermore, we provide a comprehensive summary of the established multifarious functions and mechanisms of M2-Exos miRNAs in tissue injury repair across different systems, while also exploring their potential applications in disease prevention, diagnosis, and clinical practice. Despite the challenges encountered, the therapeutic use of M2-Exos in clinical contexts appears promising, prompting research efforts to focus on improving the efficiency of exosome extraction and application, as well as ensuring the safety of their clinical utilization.
Collapse
Affiliation(s)
- Zhikang Zhu
- Department of Plastic Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, Zhejiang University, Yiwu, Zhejiang, People’s Republic of China
| | - Xinge Zhang
- Department of Plastic Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, Zhejiang University, Yiwu, Zhejiang, People’s Republic of China
| | - Xuran Lin
- Department of Plastic Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, Zhejiang University, Yiwu, Zhejiang, People’s Republic of China
| | - Yuechen Wang
- Department of Plastic Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, Zhejiang University, Yiwu, Zhejiang, People’s Republic of China
| | - Chunmao Han
- Department of Burns & Wound Care Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Shoujie Wang
- Department of Plastic Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, Zhejiang University, Yiwu, Zhejiang, People’s Republic of China
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
7
|
Negah SS, Moradi HR, Forouzanfar F, Sahraian MA, Faraji M. The Role of Small Extracellular Vesicles Derived from Glial Cells in the Central Nervous System under both Normal and Pathological Conditions. Neurochem Res 2025; 50:89. [PMID: 39883187 DOI: 10.1007/s11064-025-04344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/06/2025] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
In recent decades, researchers and clinicians have increasingly focused on glial cell function. One of the primary mechanisms influencing these functions is through extracellular vesicles (EVs), membrane-bound particles released by cells that are essential for intercellular communication. EVs can be broadly categorized into four main types based on their size, origin, and biogenesis: large EVs, small EVs (sEVs), autophagic EVs, and apoptotic bodies. Small EVs (sEVs) are involved in various physiological and pathological processes such as immune responses, angiogenesis, and cellular communication, primarily by transferring proteins, lipids, and nucleic acids to recipient cells. Interactions among glial cells mediated by small EVs can significantly modulate cell polarization and influence glial behavior through miRNA transfer. This communication, facilitated by small EVs in glial cells, is crucial for neuroinflammation, immune responses, and disease progression. This comprehensive review focuses on driven by glial small EVs, highlighting their roles in transporting biomolecules and modulating the functions of recipient cells. Furthermore, we provide an in-depth overview of the specific contributions of small EVs derived from three principal types of glial cells: oligodendrocytes, astrocytes, and microglia.
Collapse
Affiliation(s)
- Sajad Sahab Negah
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Moradi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ali Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Faraji
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
8
|
Wang X, Wang T, Zhu D, Wang J, Han W. From acute lung injury to cerebral ischemia: a unified concept involving intercellular communication through extracellular vesicle-associated miRNAs released by macrophages/microglia. Clin Exp Immunol 2025; 219:uxae105. [PMID: 39658101 PMCID: PMC11773807 DOI: 10.1093/cei/uxae105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/30/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
Ischemic stroke and acute lung injury are prevalent life-threatening conditions marked by intricate molecular mechanisms and elevated mortality rates. Despite evident pathophysiological distinctions, a notable similarity exists in the gene responses to tissue injury observed in both pathologies. This similarity extends to both protein-encoding RNAs and non-coding RNAs. Extracellular vesicles (EVs) are nano-scale vesicles derived through cell secretion, possessing unique advantages such as high biocompatibility, low immunogenicity, intrinsic cell targeting, and facile chemical and genetic manipulation. Importantly, miRNAs, the most prevalent non-coding RNAs, are selectively concentrated within EVs. Macrophages/microglia serve as immune defense and homeostatic cells, deriving from progenitor cells in the bone marrow. They can be classified into two contrasting types: classical proinflammatory M1 phenotype or alternative anti-inflammatory M2 phenotype. However, there exists a continuum of various intermediate phenotypes between M1 and M2, and macrophages/microglia can transition from one phenotype to another. This review will investigate recent discoveries concerning the impact of EVs derived from macrophages/microglia under various states on the progression of ischemic stroke and acute lung injury. The focus will be on the involvement of miRNAs within these vesicles. The concluding remarks of this review will underscore the clinical possibilities linked to EV-miRNAs, accentuating their potential as both biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xianbin Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Ting Wang
- Department of Radiology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Dong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Jing Wang
- Graduate School of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Weijie Han
- Department of General Surgery, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
9
|
Lu D, Sun H, Fan H, Li N, Li Y, Yin X, Fan Y, Sun H, Wang S, Xin T. Regulation of nerve cells and therapeutic potential in central nervous system injury using microglia-derived exosomes. Neuroscience 2024; 563:84-92. [PMID: 39521323 DOI: 10.1016/j.neuroscience.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
The intercellular communication within the central nervous system (CNS) is of great importance for in maintaining brain function, homeostasis, and CNS regulation. When the equilibrium of CNS is disrupted or injured, microglia are immediately activated and respond to CNS injury. Microglia-derived exosomes are capable of participating in intercellular communication within the CNS by transporting various bioactive substances, including nucleic acids, proteins, lipids, amino acids, and metabolites. Nevertheless, microglia activation is a double-edged sword. Activated microglia can coordinate the neural repair process and, conversely, can amplify tissue injury and impede CNS repair. This work reviewed the roles of exosomes derived from microglia stimulated by different environments (mainly lipopolysaccharide, interleukin-4, and other specific preconditioning) in CNS injury and their possible therapeutic potentials. This work focuses on the regulation of exosomes derived from microglia stimulated by different environments on nerve cells. Meanwhile, we summarized the molecular mechanisms by which the relevant exosomes exert regulatory effects. Exosomes, derived from microglia stimulated by different environments, regulate other nerve cells during the repair of CNS injury, having beneficial or detrimental effects on CNS repair. A comprehensive understanding of the molecular mechanisms underlying their role can provide a robust foundation for the clinical treatment of CNS injury.
Collapse
Affiliation(s)
- Dongxiao Lu
- College of Clinical Medicine, Jining Medical University, Jining 272067, China; Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Haohan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Hao Fan
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China; Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250021, China
| | - Nianlu Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Yuming Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Xianyong Yin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Yang Fan
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Hao Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China; Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China; Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China; Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250021, China; Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
10
|
Zhou W, Jiang X, Gao J. Extracellular vesicles for delivering therapeutic agents in ischemia/reperfusion injury. Asian J Pharm Sci 2024; 19:100965. [PMID: 39640057 PMCID: PMC11617990 DOI: 10.1016/j.ajps.2024.100965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/08/2024] [Accepted: 06/29/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemia/reperfusion (I/R) injury is marked by the restriction and subsequent restoration of blood supply to an organ. This process can exacerbate the initial tissue damage, leading to further disorders, disability, and even death. Extracellular vesicles (EVs) are crucial in cell communication by releasing cargo that regulates the physiological state of recipient cells. The development of EVs presents a novel avenue for delivering therapeutic agents in I/R therapy. The therapeutic potential of EVs derived from stem cells, endothelial cells, and plasma in I/R injury has been actively investigated. Therefore, this review aims to provide an overview of the pathological process of I/R injury and the biophysical properties of EVs. We noted that EVs serve as nontoxic, flexible, and multifunctional carriers for delivering therapeutic agents capable of intervening in I/R injury progression. The therapeutic efficacy of EVs can be enhanced through various engineering strategies. Improving the tropism of EVs via surface modification and modulating their contents via preconditioning are widely investigated in preclinical studies. Finally, we summarize the challenges in the production and delivery of EV-based therapy in I/R injury and discuss how it can advance. This review will encourage further exploration in developing efficient EV-based delivery systems for I/R treatment.
Collapse
Affiliation(s)
- Weihang Zhou
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
11
|
Ghosh M, Pearse DD. The Yin and Yang of Microglia-Derived Extracellular Vesicles in CNS Injury and Diseases. Cells 2024; 13:1834. [PMID: 39594583 PMCID: PMC11592485 DOI: 10.3390/cells13221834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in maintaining neural homeostasis but can also contribute to disease and injury when this state is disrupted or conversely play a pivotal role in neurorepair. One way that microglia exert their effects is through the secretion of small vesicles, microglia-derived exosomes (MGEVs). Exosomes facilitate intercellular communication through transported cargoes of proteins, lipids, RNA, and other bioactive molecules that can alter the behavior of the cells that internalize them. Under normal physiological conditions, MGEVs are essential to homeostasis, whereas the dysregulation of their production and/or alterations in their cargoes have been implicated in the pathogenesis of numerous neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), spinal cord injury (SCI), and traumatic brain injury (TBI). In contrast, MGEVs may also offer therapeutic potential by reversing inflammation or being amenable to engineering for the delivery of beneficial biologics or drugs. The effects of MGEVs are determined by the phenotypic state of the parent microglia. Exosomes from anti-inflammatory or pro-regenerative microglia support neurorepair and cell survival by delivering neurotrophic factors, anti-inflammatory mediators, and molecular chaperones. Further, MGEVs can also deliver components like mitochondrial DNA (mtDNA) and proteins to damaged neurons to enhance cellular metabolism and resilience. MGEVs derived from pro-inflammatory microglia can have detrimental effects on neural health. Their cargo often contains pro-inflammatory cytokines, molecules involved in oxidative stress, and neurotoxic proteins, which can exacerbate neuroinflammation, contribute to neuronal damage, and impair synaptic function, hindering neurorepair processes. The role of MGEVs in neurodegeneration and injury-whether beneficial or harmful-largely depends on how they modulate inflammation through the pro- and anti-inflammatory factors in their cargo, including cytokines and microRNAs. In addition, through the propagation of pathological proteins, such as amyloid-beta and alpha-synuclein, MGEVs can also contribute to disease progression in disorders such as AD and PD, or by the transfer of apoptotic or necrotic factors, they can induce neuron toxicity or trigger glial scarring during neurological injury. In this review, we have provided a comprehensive and up-to-date understanding of the molecular mechanisms underlying the multifaceted role of MGEVs in neurological injury and disease. In particular, the role that specific exosome cargoes play in various pathological conditions, either in disease progression or recovery, will be discussed. The therapeutic potential of MGEVs has been highlighted including potential engineering methodologies that have been employed to alter their cargoes or cell-selective targeting. Understanding the factors that influence the balance between beneficial and detrimental exosome signaling in the CNS is crucial for developing new therapeutic strategies for neurodegenerative diseases and neurotrauma.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
12
|
Zhu L, Ma L, Du X, Jiang Y, Gao J, Fan Z, Zheng H, Zhu J, Zhang G. M2 Microglia-Derived Exosomes Protect Against Glutamate-Induced HT22 Cell Injury via Exosomal miR-124-3p. Mol Neurobiol 2024; 61:7845-7861. [PMID: 38433165 DOI: 10.1007/s12035-024-04075-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
As one of the most serious complications of sepsis, sepsis-associated encephalopathy has not been effectively treated or prevented. Exosomes, as a new therapeutic method, play a protective role in neurodegenerative diseases, stroke and traumatic brain injury in recent years. The purpose of this study was to investigate the role of exosomes in glutamate (Glu)-induced neuronal injury, and to explore its mechanism, providing new ideas for the treatment of sepsis-associated encephalopathy. The neuron damage model induced by Glu was established, and its metabolomics was analyzed and identified. BV2 cells were induced to differentiate into M1 and M2 subtypes. After the exosomes from both M1-BV2 cells and M2-BV2 cells were collected, exosome morphological identification was performed by transmission electron microscopy and exosome-specific markers were also detected. These exosomes were then cocultured with HT22 cells. CCK-8 method and LDH kit were used to detect cell viability and toxicity. Cell apoptosis, mitochondrial membrane potential and ROS content were respectively detected by flow cytometry, JC-1 assay and DCFH-DA assay. MiR-124-3p expression level was detected by qRT-PCR and Western blot. Bioinformatics analysis and luciferase reporter assay predicted and verified the relationship between miR-124-3p and ROCK1 or ROCK2. Through metabolomics, 81 different metabolites were found, including fructose, GABA, 2, 4-diaminobutyric acid, etc. The enrichment analysis of differential metabolites showed that they were mainly enriched in glutathione metabolism, glycine and serine metabolism, and urea cycle. M2 microglia-derived exosomes could reduce the apoptosis, decrease the accumulation of ROS, restore the mitochondrial membrane potential and the anti-oxidative stress ability in HT22 cells induced by Glu. It was also found that the protective effect of miR-124-3p mimic on neurons was comparable to that of M2-EXOs. Additionally, M2-EXOs might carry miR-124-3p to target ROCK1 and ROCK2 in neurons, affecting ROCK/PTEN/AKT/mTOR signaling pathway, and then reducing Glu-induced neuronal apoptosis. M2 microglia-derived exosomes may protect HT22 cells against Glu-induced injury by transferring miR-124-3p into HT22 cells, with ROCK being a target gene for miR-124-3p.
Collapse
Affiliation(s)
- Lan Zhu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Limei Ma
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Xin Du
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Yuhao Jiang
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jiake Gao
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Zihao Fan
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Hengheng Zheng
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China
| | - Jianjun Zhu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu, 215004, People's Republic of China.
| | - Gaofeng Zhang
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, No.6 Huanghe Road, Changshu, Jiangsu, 215500, People's Republic of China.
| |
Collapse
|
13
|
Zhang Q, Liu J, Wang W, Lin W, Ahmed W, Duan W, Huang S, Zhu Z, Chen L. The role of exosomes derived from stem cells in nerve regeneration: A contribution to neurological repair. Exp Neurol 2024; 380:114882. [PMID: 39002923 DOI: 10.1016/j.expneurol.2024.114882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/27/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Stem cell-derived exosomes have gained attention in regenerative medicine for their role in encouraging nerve regeneration and potential use in treating neurological diseases. These nanosized extracellular vesicles act as carriers of bioactive molecules, facilitating intercellular communication and enhancing the regenerative process in neural tissues. This comprehensive study explores the methods by which exosomes produced from various stem cells contribute to nerve healing, with a particular emphasis on their role in angiogenesis, inflammation, and cellular signaling pathways. By examining cutting-edge developments and exploring the potential of exosomes in delivering disease-specific miRNAs and proteins, we highlight their versatility in tailoring personalized therapeutic strategies. The findings presented here highlight the potential of stem cell-produced exosomes for use in neurological diseases therapy, establishing the door for future research into exosome-based neurotherapies.
Collapse
Affiliation(s)
- Qiankun Zhang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiale Liu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wentong Lin
- Department of Orthopaedics, Chaozhou Hospital of Traditional Chinese Medicine, Chaozhou, China
| | - Waqas Ahmed
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Wenjie Duan
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Songze Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihan Zhu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Li X, Xia K, Zhong C, Chen X, Yang F, Chen L, You J. Neuroprotective effects of GPR68 against cerebral ischemia-reperfusion injury via the NF-κB/Hif-1α pathway. Brain Res Bull 2024; 216:111050. [PMID: 39147243 DOI: 10.1016/j.brainresbull.2024.111050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND G protein-coupled receptor 68 (GPR68), an orphan receptor, has emerged as a promising therapeutic target for mitigating neuronal inflammation and oxidative damage. This study explores the protective mechanisms of GPR68 in cerebral ischemia-reperfusion injury (CIRI). METHODS An in vivo middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model was established. Mice received intraperitoneal injections of Ogerin, a selective GPR68 agonist. In vitro, GPR68 was overexpressed in SH-SY5Y and HMC3 cells, and the effects of oxygen-glucose deprivation/reperfusion (OGD/R) on cell viability were assessed using real-time quantitative polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), and flow cytometry. RESULTS The expression of GPR68 was suppressed in cells subjected to OGD/R treatment, whereas its upregulation conferred protection to SH-SY5Y and HMC3 cells. In vivo, levels of GPR68 were reduced in brain tissues affected by MCAO/R, correlating with oxidative stress, inflammation, and neurological damage. Treatment with a GPR68 agonist decreased brain infarction, apoptosis, and dysregulated gene expression induced by MCAO/R. Mechanistically, GPR68 agonist treatment may inhibit the activation of the NF-κB/Hif-1α pathway, thereby reducing oxidative and inflammatory responses and enhancing protection against CIRI. CONCLUSIONS This study confirms that the GPR68/NF-κB/Hif-1α axis modulates apoptosis, inflammation, and oxidative stress in CIRI, indicating that GPR68 is a potential therapeutic target for CIRI.
Collapse
Affiliation(s)
- Xianglong Li
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China; Neurosurgical Clinical Research Center and Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, PR China; Laboratory of Neurological Diseases and Brain Functions, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Kaiguo Xia
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Chuanhong Zhong
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Xiangzhou Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Fubing Yang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China; Neurosurgical Clinical Research Center and Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, PR China; Laboratory of Neurological Diseases and Brain Functions, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China.
| | - Jian You
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China; Neurosurgical Clinical Research Center and Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, PR China; Laboratory of Neurological Diseases and Brain Functions, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China.
| |
Collapse
|
15
|
Liu Y, Yang P, Wang J, Peng W, Zhao J, Wang Z. MiRNA Regulates Ferroptosis in Cardiovascular and Cerebrovascular Diseases. DNA Cell Biol 2024; 43:492-509. [PMID: 39417991 DOI: 10.1089/dna.2024.0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) significantly contribute to global mortality and morbidity due to their complex pathogenesis involving multiple biological processes. Ferroptosis is an important physiological process in CCVDs, manifested by an abnormal increase in intracellular iron concentration. MiRNAs, a key class of noncoding RNA molecules, are crucial in regulating CCVDs through pathways like glutathione-glutathione peroxidase 4, glutamate/cystine transport, iron metabolism, lipid metabolism, and other oxidative stress pathways. This article summarizes the progress of miRNAs' regulation on CCVDs, aiming to provide insights for the diagnosis and treatment of CCVDs.
Collapse
Affiliation(s)
- Yiman Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Peijuan Yang
- Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Jingjing Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Wu Peng
- Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Jinli Zhao
- Emergency Department, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
16
|
Ryou MG, Burton S. Intermittent hypoxic training - derived exosomes in stroke rehabilitation. Front Integr Neurosci 2024; 18:1475234. [PMID: 39323911 PMCID: PMC11422222 DOI: 10.3389/fnint.2024.1475234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 08/19/2024] [Indexed: 09/27/2024] Open
Abstract
Ischemic stroke is the fourth leading cause of adult disability in the US, and it is a huge social burden all over the world. However, the efficient treatment of ischemic stroke is not available. An apparent reason for failing to find or develop an intervention for ischemic stroke is contributed to the tight blood-brain barrier (BBB). The unique characteristics of exosomes that can traverse BBB have been highlighted among researchers investigating interventions for ischemic stroke conditions. Additionally, intermittent hypoxic training has been considered a potential intervention in the treatment or rehabilitation process of ischemic stroke patients. In this mini-review, we are going to review the possibility of applying exosomes produced by a subject who does intermittent hypoxic conditioning in a treatment program for ischemic stroke.
Collapse
Affiliation(s)
- Myoung-Gwi Ryou
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Science, Tarleton State University, Fort Worth, TX, United States
| | - Summer Burton
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Science, Tarleton State University, Fort Worth, TX, United States
| |
Collapse
|
17
|
Fang X, Zhou D, Wang X, Ma Y, Zhong G, Jing S, Huang S, Wang Q. Exosomes: A Cellular Communication Medium That Has Multiple Effects On Brain Diseases. Mol Neurobiol 2024; 61:6864-6892. [PMID: 38356095 DOI: 10.1007/s12035-024-03957-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Exosomes, as membranous vesicles generated by multiple cell types and secreted to extracellular space, play a crucial role in a range of brain injury-related brain disorders by transporting diverse proteins, RNA, DNA fragments, and other functional substances. The nervous system's pathogenic mechanisms are complicated, involving pathological processes like as inflammation, apoptosis, oxidative stress, and autophagy, all of which result in blood-brain barrier damage, cognitive impairment, and even loss of normal motor function. Exosomes have been linked to the incidence and progression of brain disorders in recent research. As a result, a thorough knowledge of the interaction between exosomes and brain diseases may lead to the development of more effective therapeutic techniques that may be implemented in the clinic. The potential role of exosomes in brain diseases and the crosstalk between exosomes and other pathogenic processes were discussed in this paper. Simultaneously, we noted the delicate events in which exosomes as a media allow the brain to communicate with other tissues and organs in physiology and disease, and compiled a list of natural compounds that modulate exosomes, in order to further improve our understanding of exosomes and propose new ideas for treating brain disorders.
Collapse
Affiliation(s)
- Xiaoling Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Dishu Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Xinyue Wang
- Department of Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510405, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 510405, Guangzhou, China
| | - Yujie Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Shangwen Jing
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China
| | - Shuiqing Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China.
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong Province, China.
| |
Collapse
|
18
|
Nouri Z, Barfar A, Perseh S, Motasadizadeh H, Maghsoudian S, Fatahi Y, Nouri K, Yektakasmaei MP, Dinarvand R, Atyabi F. Exosomes as therapeutic and drug delivery vehicle for neurodegenerative diseases. J Nanobiotechnology 2024; 22:463. [PMID: 39095888 PMCID: PMC11297769 DOI: 10.1186/s12951-024-02681-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/30/2024] [Indexed: 08/04/2024] Open
Abstract
Neurodegenerative disorders are complex, progressive, and life-threatening. They cause mortality and disability for millions of people worldwide. Appropriate treatment for neurodegenerative diseases (NDs) is still clinically lacking due to the presence of the blood-brain barrier (BBB). Developing an effective transport system that can cross the BBB and enhance the therapeutic effect of neuroprotective agents has been a major challenge for NDs. Exosomes are endogenous nano-sized vesicles that naturally carry biomolecular cargoes. Many studies have indicated that exosome content, particularly microRNAs (miRNAs), possess biological activities by targeting several signaling pathways involved in apoptosis, inflammation, autophagy, and oxidative stress. Exosome content can influence cellular function in healthy or pathological ways. Furthermore, since exosomes reflect the features of the parental cells, their cargoes offer opportunities for early diagnosis and therapeutic intervention of diseases. Exosomes have unique characteristics that make them ideal for delivering drugs directly to the brain. These characteristics include the ability to pass through the BBB, biocompatibility, stability, and innate targeting properties. This review emphasizes the role of exosomes in alleviating NDs and discusses the associated signaling pathways and molecular mechanisms. Furthermore, the unique biological features of exosomes, making them a promising natural transporter for delivering various medications to the brain to combat several NDs, are also discussed.
Collapse
Affiliation(s)
- Zeinab Nouri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashkan Barfar
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahra Perseh
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Motasadizadeh
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samane Maghsoudian
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Keyvan Nouri
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Rassoul Dinarvand
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Poinsot V, Pizzinat N, Ong-Meang V. Engineered and Mimicked Extracellular Nanovesicles for Therapeutic Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:639. [PMID: 38607173 PMCID: PMC11013861 DOI: 10.3390/nano14070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Exosomes are spherical extracellular nanovesicles with an endosomal origin and unilamellar lipid-bilayer structure with sizes ranging from 30 to 100 nm. They contain a large range of proteins, lipids, and nucleic acid species, depending on the state and origin of the extracellular vesicle (EV)-secreting cell. EVs' function is to encapsulate part of the EV-producing cell content, to transport it through biological fluids to a targeted recipient, and to deliver their cargos specifically within the aimed recipient cells. Therefore, exosomes are considered to be potential biological drug-delivery systems that can stably deliver their cargo into targeted cells. Various cell-derived exosomes are produced for medical issues, but their use for therapeutic purposes still faces several problems. Some of these difficulties can be avoided by resorting to hemisynthetic approaches. We highlight here the uses of alternative exosome-mimes involving cell-membrane coatings on artificial nanocarriers or the hybridization between exosomes and liposomes. We also detail the drug-loading strategies deployed to make them drug-carrier systems and summarize the ongoing clinical trials involving exosomes or exosome-like structures. Finally, we summarize the open questions before considering exosome-like disposals for confident therapeutic delivery.
Collapse
Affiliation(s)
- Verena Poinsot
- Inserm, CNRS, Faculté de Santé, Université Toulouse III—Paul Sabatier, I2MC U1297, 31432 Toulouse, France; (N.P.); (V.O.-M.)
| | | | | |
Collapse
|
20
|
Yu Y, Liao X, Xie X, Li Q, Chen X, Liu R. The role of neuroglial cells communication in ischemic stroke. Brain Res Bull 2024; 209:110910. [PMID: 38423190 DOI: 10.1016/j.brainresbull.2024.110910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
Ischemic stroke is one of the leading causes of death and disability globally, but its treatment options are limited due to therapeutic window and reperfusion injury constraints. Microglia, astrocytes, and oligodendrocytes are the major components of the neurovascular unit, and there is substantial evidence suggesting their contributions to maintaining homeostasis in the central nervous system. Neuroglial cells participate in neuronal physiological functions and the repair of damaged neurons through various communication methods, including gap junctions, chemical signaling, and extracellular vesicles, in conjunction with other components of the neurovascular unit. Ischemia-induced microglia and astrocytes polarize into "M1/M2" and "A1/A2" phenotypes and exert neurotoxic or neuroprotective effects by releasing soluble factors, secreting extracellular vesicles, and forming syncytia networks in the acute (<72 h), subacute (>72 h), and chronic phases (>6 weeks). Apoptosis of oligodendrocytes due to ischemic hypoxia leads to white matter injury, causing long-term cognitive dysfunction, and promoting oligodendrogenesis is a crucial direction for achieving functional recovery in ischemic stroke. In this article, we summarize the cellular interactions following cerebral ischemia, analyze the roles of neuroglial cells through gap junctions, chemical signaling, and extracellular vesicles in different stages of ischemic stroke, and further explore strategies for intervening in ischemic stroke.
Collapse
Affiliation(s)
- Yunling Yu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xinglan Liao
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xinyu Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Qihua Li
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xuehong Chen
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Ruizhen Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
21
|
Singh G, Mehra A, Arora S, Gugulothu D, Vora LK, Prasad R, Khatri DK. Exosome-mediated delivery and regulation in neurological disease progression. Int J Biol Macromol 2024; 264:130728. [PMID: 38467209 DOI: 10.1016/j.ijbiomac.2024.130728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/13/2024]
Abstract
Exosomes (EXOs), membranous structures originating from diverse biological sources, have recently seized the attention of researchers due to their theranostic potential for neurological diseases. Released actively by various cells, including stem cells, adipose tissue, and immune cells, EXOs wield substantial regulatory influence over the intricate landscape of neurological complications, exhibiting both positive and negative modulatory effects. In AD, EXOs play a pivotal role in disseminating and breaking down amyloid-β protein. Moreover, EXOs derived from mesenchymal stem cells showcase a remarkable capacity to mitigate pro-inflammatory phenotypes by regulating miRNAs in neurodegenerative diseases. These vesicles possess the unique ability to traverse the blood-brain barrier, governing the aggregation of mutant huntingtin protein. Understanding the exosomal functions within the CNS holds significant promise for enhancing treatment efficacy in neurological diseases. This review intricately examines the regulatory mechanisms involving EXOs in neurological disease development, highlighting therapeutic prospects and exploring their utility in exosome-based nanomedicine for various neurological complications. Additionally, the review highlights the challenges associated with drug delivery to the brain, emphasizing the complexities inherent in this critical aspect of neurotherapeutics.
Collapse
Affiliation(s)
- Gurpreet Singh
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, India
| | - Ankit Mehra
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, India
| | - Sanchit Arora
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), M.B. Road, Pushp Vihar, Sector-3, New Delhi 110017, India
| | - Dalapathi Gugulothu
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), M.B. Road, Pushp Vihar, Sector-3, New Delhi 110017, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK.
| | - Renuka Prasad
- Department of Anatomy, Korea University College of Medicine, Moonsuk Medical Research Building, 516, 5th floor, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Dharmendra Kumar Khatri
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, India; Department of Pharmacology, Shobhaben Pratapbai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-University, Mumbai 400056, India.
| |
Collapse
|
22
|
Hu S, Feng L, Yang Z, Fan X, Gao H, Yang T. A recognition of exosomes as regulators of epigenetic mechanisms in central nervous system diseases. Front Mol Neurosci 2024; 17:1370449. [PMID: 38528957 PMCID: PMC10962328 DOI: 10.3389/fnmol.2024.1370449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/28/2024] [Indexed: 03/27/2024] Open
Abstract
Exosomes, vesicular structures originating from cells, participate in the conveyance of proteins and nucleic acids. Presently, the centrality of epigenetic modifications in neurological disorders is widely acknowledged. Exosomes exert influence over various epigenetic phenomena, thereby modulating post-transcriptional regulatory processes contingent upon their constituent makeup. Consequently, the heightened attention directed toward exosomes as instigators of epigenetic alterations has burgeoned in recent years. Notably, exosomes serve as vehicles for delivering methyltransferases to recipient cells. More significantly, non-coding RNAs, particularly microRNAs (miRNAs), represent pivotal contents within exosomes, wielding the capacity to influence the expression of diverse factors within the cerebral milieu. The transfer of these exosomal contents amidst brain cells, encompassing neuronal cells and microglia, assumes a critical role in the genesis and progression of neurological disorders, also, this role is not limited to neurological disorders, it may deal with any human disease, such as cancer, and cardiovascular diseases. This review will concentrate on elucidating the regulation of exosome-induced epigenetic events and its subsequent ramifications for neurological diseases. A more profound comprehension of the involvement of exosome-mediated epigenetic regulation in neurological disorders contributes to a heightened awareness of the etiology and advancement of cerebral afflictions.
Collapse
Affiliation(s)
- Shunxin Hu
- Shandong First Medical University, Tai'an, China
| | - Lei Feng
- Jining First People's Hospital, Jining, China
| | | | - Xuechen Fan
- Jining First People's Hospital, Jining, China
| | | | | |
Collapse
|
23
|
Li Z, Pang Y, Hou L, Xing X, Yu F, Gao M, Wang J, Li X, Zhang L, Xiao Y. Exosomal OIP5-AS1 attenuates cerebral ischemia-reperfusion injury by negatively regulating TXNIP protein stability and inhibiting neuronal pyroptosis. Int Immunopharmacol 2024; 127:111310. [PMID: 38103409 DOI: 10.1016/j.intimp.2023.111310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/15/2023] [Accepted: 11/26/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) can cause neuronal apoptosis and lead to irreversible brain injury. Numerous lncRNAs have been reported to play important roles in CIRI, but it is unclear whether these lncRNAs can function through exosomes. METHODS In this study, we utilized the middle cerebral artery occlusion/reperfusion (MCAO/R) animal model and the oxygen-glucose deprivation/ reoxygenation (OGD/R) cell model. RNA sequencing was performed to screen for differentially expressed lncRNAs in M2 microglia-derived exosomes (M2-Exos). RNA pull-down, RNA immunoprecipitation, co-immunoprecipitation and ubiquitination assays were used to explore the molecular mechanism of OIP5-AS1 in alleviating CIRI. RESULTS M2-Exos could alleviate nerve injury and pyroptosis after CIRI in vitro and in vivo. OIP5-AS1 was found to be significantly up-regulated in M2-Exos and down-regulated in OGD/R neurons, MCAO/R mice and ischemic stroke patients. In MCAO/R mice, OIP5-AS1 could reduce cerebral infarct size, cerebral edema and mNSS scores, and inhibit the expression levels of pyroptosis-related proteins in brain tissue. TXNIP was confirmed to be a reliable binding protein of OIP5-AS1. OIP5-AS1 overexpression significantly attenuated MCAO/R-induced upregulation of TXNIP at the protein level, but not at the mRNA level. OIP5-AS1 promoted the TXNIP degradation process and increased the ubiquitination of TXNIP. ITCH could bind to TXNIP. ITCH overexpression or knockdown did not alter the mRNA level of TXNIP, but negatively regulated TXNIP expression at the protein level. ITCH accelerated the degradation and ubiquitination of TXNIP, which could be attenuated by OIP5-AS1 knockdown. OIP5-AS1 could improve neuronal damage and inhibit neuronal pyroptosis through TXNIP. CONCLUSIONS M2-Exo-derived OIP5-AS1 can induce TXNIP ubiquitination and degradation by recruiting ITCH, negatively regulate TXNIP protein stability, inhibit neuronal pyroptosis, and attenuate CIRI.
Collapse
Affiliation(s)
- Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China; Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan City, Shandong Province 250063, China
| | - Yuejiu Pang
- Department of Healthcare Neurology, Provincial Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province 250021, China
| | - Lei Hou
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Fuhua Yu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Mingxu Gao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China.
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China.
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China.
| |
Collapse
|
24
|
Ciaccio AM, Tuttolomondo A. Exosomal miRNAs as Biomarkers of Ischemic Stroke. Brain Sci 2023; 13:1647. [PMID: 38137095 PMCID: PMC10741776 DOI: 10.3390/brainsci13121647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/08/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are small lipid bilayer membrane particles released from all living cells into the extracellular environment. They carry several molecules and have a critical role in cell-cell communication under physiological and pathological conditions. In recent decades, exosomes, and especially their cargo, have emerged as a promising tool for several clinical conditions. However, the literature has become increasingly unambiguous in defining the role of exosomes in chronic cerebrovascular diseases. Because they can pass through the blood-brain barrier, they have great potential to reflect intracerebral changes. They can, thus, provide valuable insight into the mechanisms of central nervous system diseases. The purpose of this review is to describe the literature on the role of exosomal miRNA, which represents the most widely investigated exosomal biomarker, in strokes. First, we provide an overview of exosomes, from biology to isolation and characterization. Then, we describe the relationship between exosomes and stroke pathogenesis. Finally, we summarize the human studies evaluating exosomal miRNA biomarkers of stroke. Although the collective literature supports the potential use of exosomal miRNA as biomarkers of ischemic stroke, there are still several limitations hampering their introduction into clinical practice.
Collapse
Affiliation(s)
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care Ward, Regional Reference Center for Diagnosis and Treatment of Anderson-Fabry Disease, Department of Health Promotion, Maternal and Child Health, Internal Medicine, and Specialty Excellence “G. D’Alessandro” (PROMISE), University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
25
|
Wang Y, Liu W, Geng P, Du W, Guo C, Wang Q, Zheng GQ, Jin X. Role of Crosstalk between Glial Cells and Immune Cells in Blood-Brain Barrier Damage and Protection after Acute Ischemic Stroke. Aging Dis 2023; 15:2507-2525. [PMID: 37962453 PMCID: PMC11567273 DOI: 10.14336/ad.2023.1010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
Blood-brain barrier (BBB) damage is the main pathological basis for acute ischemic stroke (AIS)-induced cerebral vasogenic edema and hemorrhagic transformation (HT). Glial cells, including microglia, astrocytes, and oligodendrocyte precursor cells (OPCs)/oligodendrocytes (OLs) play critical roles in BBB damage and protection. Recent evidence indicates that immune cells also have an important role in BBB damage, vasogenic edema and HT. Therefore, regulating the crosstalk between glial cells and immune cells would hold the promise to alleviate AIS-induced BBB damage. In this review, we first introduce the roles of glia cells, pericytes, and crosstalk between glial cells in the damage and protection of BBB after AIS, emphasizing the polarization, inflammatory response and crosstalk between microglia, astrocytes, and other glia cells. We then describe the role of glial cell-derived exosomes in the damage and protection of BBB after AIS. Next, we specifically discuss the crosstalk between glial cells and immune cells after AIS. Finally, we propose that glial cells could be a potential target for alleviating BBB damage after AIS and we discuss some molecular targets and potential strategies to alleviate BBB damage by regulating glial cells after AIS.
Collapse
Affiliation(s)
- Yihui Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Wencao Liu
- Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| | - Guo-qing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
26
|
Chen Y, Pang J, Ye L, Zhang Z, Lin S, Lin N, Lee TH, Liu H. Disorders of the central nervous system: Insights from Notch and Nrf2 signaling. Biomed Pharmacother 2023; 166:115383. [PMID: 37643483 DOI: 10.1016/j.biopha.2023.115383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
The functional complexity of the central nervous system (CNS) is unparalleled in living organisms. It arises from neural crest-derived cells that migrate by the exact route, leading to the formation of a complex network of neurons and glial cells. Recent studies have shown that novel crosstalk exists between the Notch1 and Nrf2 pathways and is associated with many neurological diseases. The Notch1-Nrf2 axis may act on nervous system development, and the molecular mechanism has recently been reported. In this review, we summarize the essential structure and function of the CNS. The significance of interactions between signaling pathways and between developmental processes like proliferation, apoptosis and migration in ensuring the correct development of the CNS is also presented. We primarily focus on research concerning possible mechanism of interaction between Notch1 and Nrf2 and the functions of Notch1-Nrf2 in neurons. There may be a direct interaction between Notch1 and NRF2, which is closely related to the crosstalk that occurs between them. The significance and potential applications of the Notch1-Nrf2 axis in abnormal development of the nervous system are been highlighten. We also discuss the molecular mechanisms by which the Notch1-Nrf2 axis controls the apoptosis, antioxidant pathway and differentiation of neurons to modulate the development of the nervous system. This information will lead to a better understanding of Notch1-Nrf2 axis signaling pathways in the nervous system and may facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Yuwen Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jiao Pang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Lu Ye
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Zhentao Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Suijin Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Na Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
27
|
Xin W, Pan Y, Wei W, Tatenhorst L, Graf I, Popa-Wagner A, Gerner ST, Huber S, Kilic E, Hermann DM, Bähr M, Huttner HB, Doeppner TR. Preconditioned extracellular vesicles from hypoxic microglia reduce poststroke AQP4 depolarization, disturbed cerebrospinal fluid flow, astrogliosis, and neuroinflammation. Theranostics 2023; 13:4197-4216. [PMID: 37554272 PMCID: PMC10405850 DOI: 10.7150/thno.84059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/06/2023] [Indexed: 08/10/2023] Open
Abstract
Background: Stroke stimulates reactive astrogliosis, aquaporin 4 (AQP4) depolarization and neuroinflammation. Preconditioned extracellular vesicles (EVs) from microglia exposed to hypoxia, in turn, reduce poststroke brain injury. Nevertheless, the underlying mechanisms of such effects are elusive, especially with regards to inflammation, AQP4 polarization, and cerebrospinal fluid (CSF) flow. Methods: Primary microglia and astrocytes were exposed to oxygen-glucose deprivation (OGD) injury. For analyzing the role of AQP4 expression patterns under hypoxic conditions, a co-culture model of astrocytes and microglia was established. Further studies applied a stroke model, where some mice also received an intracisternal tracer infusion of rhodamine B. As such, these in vivo studies involved the analysis of AQP4 polarization, CSF flow, astrogliosis, and neuroinflammation as well as ischemia-induced brain injury. Results: Preconditioned EVs decreased periinfarct AQP4 depolarization, brain edema, astrogliosis, and inflammation in stroke mice. Likewise, EVs promoted postischemic CSF flow and cerebral blood perfusion, and neurological recovery. Under in vitro conditions, hypoxia stimulated M2 microglia polarization, whereas EVs augmented M2 microglia polarization and repressed M1 microglia polarization even further. In line with this, astrocytes displayed upregulated AQP4 clustering and proinflammatory cytokine levels when exposed to OGD, which was reversed by preconditioned EVs. Reduced AQP4 depolarization due to EVs, however, was not a consequence of unspecific inflammatory regulation, since LPS-induced inflammation in co-culture models of astrocytes and microglia did not result in altered AQP4 expression patterns in astrocytes. Conclusions: These findings show that hypoxic microglia may participate in protecting against stroke-induced brain damage by regulating poststroke inflammation, astrogliosis, AQP4 depolarization, and CSF flow due to EV release.
Collapse
Affiliation(s)
- Wenqiang Xin
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Yongli Pan
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Irina Graf
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Aurel Popa-Wagner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stefan T Gerner
- Department of Neurology, University of Giessen Medical School, Giessen, Germany
| | - Sabine Huber
- Department of Neurology, University of Giessen Medical School, Giessen, Germany
| | - Ertugrul Kilic
- Department of Physiology, Istanbul Medeniyet University, Istanbul, Turkey
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mathias Bähr
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Hagen B Huttner
- Department of Neurology, University of Giessen Medical School, Giessen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
- Department of Neurology, University of Giessen Medical School, Giessen, Germany
- Department of Anatomy and Cell Biology, Medical University of Varna, Varna, Bulgaria
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
- Research Institute for Health Sciences and Technologies (SABITA), Medipol University, Istanbul, Turkey
| |
Collapse
|
28
|
Wang L, Wei X. Exosome-based crosstalk in glaucoma pathogenesis: a focus on oxidative stress and neuroinflammation. Front Immunol 2023; 14:1202704. [PMID: 37529047 PMCID: PMC10388248 DOI: 10.3389/fimmu.2023.1202704] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/03/2023] [Indexed: 08/03/2023] Open
Abstract
Exosomes are membrane-bound tiny particles that are released by all live cells that contain multiple signal molecules and extensively participate in numerous normal physical activities and pathologies. In glaucoma, the crucial role of exosome-based crosstalk has been primarily revealed in animal models and ex vivo cell studies in the recent decade. In the aqueous drainage system, exosomes derived from non-pigment ciliary epithelium act in an endocrine manner and specifically regulate the function of the trabecular meshwork to cope with persistent oxidative stress challenges. In the retina, a more complicated regulatory network among microglia, retinal neurons, retinal ganglial cells, retinal pigment epithelium, and other immune effector cells by exosomes are responsible for the elaborate modulation of tissue homeostasis under physical state and the widespread propagation of neuroinflammation and its consequent neurodegeneration in glaucoma pathogenesis. Accumulating evidence indicates that exosome-based crosstalk depends on numerous factors, including the specific cargos they carried (particularly micro RNA), concentration, size, and ionization potentials, which largely remain elusive. In this narrative review, we summarize the latest research focus of exosome-based crosstalk in glaucoma pathogenesis, the current research progress of exosome-based therapy for glaucoma and provide in-depth perspectives on its current research gap.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Ophthalmology, ShangjinNanfu Hospital, Chengdu, China
| |
Collapse
|
29
|
Xie K, Mo Y, Yue E, Shi N, Liu K. Exosomes derived from M2-type microglia ameliorate oxygen-glucose deprivation/reoxygenation-induced HT22 cell injury by regulating miR-124-3p/NCOA4-mediated ferroptosis. Heliyon 2023; 9:e17592. [PMID: 37424595 PMCID: PMC10328844 DOI: 10.1016/j.heliyon.2023.e17592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/11/2023] Open
Abstract
Background Although it has been reported that miRNA carried by M2 microglial exosomes protects neurons from ischemia-reperfusion brain injury, the mechanism of action remains poorly understood. This study aimed to explore the miRNA signaling pathway by which M2-type microglia-derived exosomes (M2-exosomes) ameliorate oxygen-glucose deprivation/reoxygenation (OGD/R)-induced cytotoxicity in HT22 cells. Methods BV2 microglia were induced by M2 polarization. Then, M2-exosomes were identified via transmission electron microscopy and special biomarker detection and co-cultured with HT22 cells. Cell proliferation was evaluated using the Cell Counting Kit-8 (CCK-8) assay. Intracellular concentrations of reactive oxygen species (ROS), Fe2+, glutathione (GSH), and malondialdehyde (MDA) were determined using dichlorofluorescein fluorescence and biochemical determination. miR-124-3p levels were determined using qRT-PCR, and protein expressions were examined via western blotting. Results OGD/R suppressed the proliferation and induced the accumulation of Fe2+, ROS, and MDA and reduction of GSH in mouse HT22 cells, suggesting ferroptosis of HT22 cells. OGD/R-induced changes in the above mentioned indexes was ameliorated by M2-exosomes but restored by the exosome inhibitor GW4869. M2-exosomes with (mimic-exo) or without miR-124-3p (inhibitor-exo) promoted and suppressed proliferation and ferroptosis-associated indexes of HT22 cells, respectively. Moreover, mimic-exo and inhibitor-exo inhibited and enhanced NCOA4 expression in HT22 cells, respectively. NCOA4 overexpression reversed the protective effects of miR-124-3p mimic-exo in OGD/R-conditioned cells. NCOA4 was targeted and regulated by miR-124-3p. Conclusions M2-exosome protects HT22 cells against OGD/R-induced ferroptosis injury by transferring miR-124-3p and NCOA4 into HT22 cells, with the latter being a target gene for miR-124-3p.
Collapse
Affiliation(s)
- Ke Xie
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Yun Mo
- Department of Neurology, Guizhou Medical University, China
| | - Erli Yue
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Nan Shi
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Kangyong Liu
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| |
Collapse
|
30
|
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. Int J Mol Sci 2023; 24:10802. [PMID: 37445979 DOI: 10.3390/ijms241310802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
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.
Collapse
Affiliation(s)
- Mihai Ruscu
- Doctoral School, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
| | - Andreea Cercel
- Doctoral School, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
| | - Ertugrul Kilic
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul 34214, Turkey
| | - Bogdan Catalin
- Doctoral School, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
| | - Andrei Gresita
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 115680-8000, USA
| | - Dirk M Hermann
- Doctoral School, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
| | - Aurel Popa-Wagner
- Doctoral School, University of Medicine and Pharmacy Craiova, 200349 Craiova, Romania
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
31
|
Wawrzkiewicz-Jałowiecka A, Lalik A, Lukasiak A, Richter-Laskowska M, Trybek P, Ejfler M, Opałka M, Wardejn S, Delfino DV. Potassium Channels, Glucose Metabolism and Glycosylation in Cancer Cells. Int J Mol Sci 2023; 24:ijms24097942. [PMID: 37175655 PMCID: PMC10178682 DOI: 10.3390/ijms24097942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Potassium channels emerge as one of the crucial groups of proteins that shape the biology of cancer cells. Their involvement in processes like cell growth, migration, or electric signaling, seems obvious. However, the relationship between the function of K+ channels, glucose metabolism, and cancer glycome appears much more intriguing. Among the typical hallmarks of cancer, one can mention the switch to aerobic glycolysis as the most favorable mechanism for glucose metabolism and glycome alterations. This review outlines the interconnections between the expression and activity of potassium channels, carbohydrate metabolism, and altered glycosylation in cancer cells, which have not been broadly discussed in the literature hitherto. Moreover, we propose the potential mediators for the described relations (e.g., enzymes, microRNAs) and the novel promising directions (e.g., glycans-orinented drugs) for further research.
Collapse
Affiliation(s)
- Agata Wawrzkiewicz-Jałowiecka
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Anna Lalik
- Department of Systems Biology and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Agnieszka Lukasiak
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Monika Richter-Laskowska
- The Centre for Biomedical Engineering, Łukasiewicz Research Network-Krakow Institute of Technology, 30-418 Krakow, Poland
| | - Paulina Trybek
- Institute of Physics, University of Silesia in Katowice, 41-500 Chorzów, Poland
| | - Maciej Ejfler
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Maciej Opałka
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Sonia Wardejn
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Domenico V Delfino
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
| |
Collapse
|
32
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
33
|
Bai Y, Ren H, Bian L, Zhou Y, Wang X, Xiong Z, Liu Z, Han B, Yao H. Regulation of Glial Function by Noncoding RNA in Central Nervous System Disease. Neurosci Bull 2023; 39:440-452. [PMID: 36161582 PMCID: PMC10043107 DOI: 10.1007/s12264-022-00950-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/24/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are a class of functional RNAs that play critical roles in different diseases. NcRNAs include microRNAs, long ncRNAs, and circular RNAs. They are highly expressed in the brain and are involved in the regulation of physiological and pathophysiological processes of central nervous system (CNS) diseases. Mounting evidence indicates that ncRNAs play key roles in CNS diseases. Further elucidating the mechanisms of ncRNA underlying the process of regulating glial function that may lead to the identification of novel therapeutic targets for CNS diseases.
Collapse
Affiliation(s)
- Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Hui Ren
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Liang Bian
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - You Zhou
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xinping Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhongli Xiong
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ziqi Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Bing Han
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Honghong Yao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory, School of Medicine, Southeast University, Nanjing, 210009, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
34
|
Abdel Mageed SS, Doghish AS, Ismail A, El-Husseiny AA, Fawzi SF, Mahmoud AMA, El-Mahdy HA. The role of miRNAs in insulin resistance and diabetic macrovascular complications - A review. Int J Biol Macromol 2023; 230:123189. [PMID: 36623613 DOI: 10.1016/j.ijbiomac.2023.123189] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023]
Abstract
Diabetes is the most prevalent metabolic disturbance disease and has been regarded globally as one of the principal causes of mortality. Diabetes is accompanied by several macrovascular complications, including stroke, coronary artery disease (CAD), and cardiomyopathy as a consequence of atherosclerosis. The onset of type 2 diabetes is closely related to insulin resistance (IR). miRNAs have been linked to various metabolic processes, including glucose homeostasis, regulation of lipid metabolism, gluconeogenesis, adipogenesis, glucose transporter type 4 expression, insulin sensitivity, and signaling. Consequently, miRNA dysregulation mediates IR in some target organs, comprising liver, muscle, and adipose tissue. Moreover, miRNAs are crucial in developing diabetes and its associated macrovascular complications through their roles in several signaling pathways implicated in inflammation, apoptosis, cellular survival and migration, the proliferation of vascular smooth muscle cells, neurogenesis, angiogenesis, autophagy, oxidative stress, cardiac remodeling, and fibrosis. Therefore, the purpose of this review is to clarify the role of miRNAs in hepatic, muscle, and adipose tissue IR and explain their roles in the pathogenesis of macrovascular diabetic complications, including stroke, CAD, and cardiomyopathy. Also, explain their roles in gestational diabetes mellitus (GDM). Besides, this review discusses the latest updates on the alteration of miRNA expression in diabetic macrovascular complications.
Collapse
Affiliation(s)
- Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt
| | - Sylvia F Fawzi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Abdulla M A Mahmoud
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt
| |
Collapse
|
35
|
Rehman FU, Liu Y, Zheng M, Shi B. Exosomes based strategies for brain drug delivery. Biomaterials 2023; 293:121949. [PMID: 36525706 DOI: 10.1016/j.biomaterials.2022.121949] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 11/12/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Exosome application has emerged as a promising nanotechnology discipline for various diseases therapeutics and diagnoses. Owing to the natural properties of efficient drug delivery, higher biocompatibility, facile traversing of physiological barriers, and subtle side effects, exosomes shorten their way to clinical translation. Exosomes are nanoscale membrane-bound vesicles primarily involved in intercellular communication and exhibit natural blood-brain barrier (BBB) traversing ability, which enables their application as drug delivery vehicles for brain diseases treatment. Herein, we highlight recent exosome-based drug delivery endeavors for neurodegenerative diseases and brain cancer therapy, summarize the obstacles and future directions in clinical translation.
Collapse
Affiliation(s)
- Fawad Ur Rehman
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China; Centre for Regenerative Medicine and Stem Cells Research, The Aga Khan University, Stadium Road, Karachi, 74800, Pakistan
| | - Yang Liu
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China.
| | - Bingyang Shi
- Henan-Macquire International Joint Center for Biomedical Innovations, School of Life Sciences, Henan University, JinMing Avenue, Kaifeng, 475004 PR China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan 475004, China; Department of Biomedical Sciences Faculty of Medicine and Health Sciences Macquarie University Sydney, NSW, 2109, Australia.
| |
Collapse
|
36
|
Exosomes as biomarkers and therapeutic measures for ischemic stroke. Eur J Pharmacol 2023; 939:175477. [PMID: 36543286 DOI: 10.1016/j.ejphar.2022.175477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/01/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Ischemic stroke (IS) is the leading cause of long-term disability in the world and characterized by high morbidity, recurrence, complications, and mortality. Due to the lack of early diagnostic indicators, limited therapeutic measures and inadequate prognostic indicators, the diagnosis and treatment of IS remains a particular challenge at present. It has recently been reported that exosomes (EXOs) play a significant role in the pathogenesis and treatment of IS. The purpose of this paper is to probe the role of EXOs in diagnostic biomarkers and therapeutic measures for IS and to provide innovative ideas for improving the prognosis of IS.
Collapse
|
37
|
Gambardella J, Kansakar U, Sardu C, Messina V, Jankauskas SS, Marfella R, Maggi P, Wang X, Mone P, Paolisso G, Sorriento D, Santulli G. Exosomal miR-145 and miR-885 Regulate Thrombosis in COVID-19. J Pharmacol Exp Ther 2023; 384:109-115. [PMID: 35772782 PMCID: PMC9827505 DOI: 10.1124/jpet.122.001209] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 01/13/2023] Open
Abstract
We hypothesized that exosomal microRNAs could be implied in the pathogenesis of thromboembolic complications in coronavirus disease 2019 (COVID-19). We isolated circulating exosomes from patients with COVID-19, and then we divided our population in two arms based on the D-dimer level on hospital admission. We observed that exosomal miR-145 and miR-885 significantly correlate with D-dimer levels. Moreover, we demonstrate that human endothelial cells express the main cofactors needed for the internalization of the "Severe acute respiratory syndrome coronavirus 2" (SARS-CoV-2), including angiotensin converting enzyme 2, transmembrane protease serine 2, and CD-147. Interestingly, human endothelial cells treated with serum from COVID-19 patients release significantly less miR-145 and miR-885, exhibit increased apoptosis, and display significantly impaired angiogenetic properties compared with cells treated with non-COVID-19 serum. Taken together, our data indicate that exosomal miR-145 and miR-885 are essential in modulating thromboembolic events in COVID-19. SIGNIFICANCE STATEMENT: This work demonstrates for the first time that two specific microRNAs (namely miR-145 and miR-885) contained in circulating exosomes are functionally involved in thromboembolic events in COVID-19. These findings are especially relevant to the general audience when considering the emerging prominence of post-acute sequelae of COVID-19 systemic manifestations known as Long COVID.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Urna Kansakar
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Celestino Sardu
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Vincenzo Messina
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Stanislovas S Jankauskas
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Raffaele Marfella
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Paolo Maggi
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Xujun Wang
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Pasquale Mone
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Giuseppe Paolisso
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Daniela Sorriento
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute (J.G., U.K., S.S.J., X.W., P.Mo.) and Department of Molecular Pharmacology, Einstein-Sinai Diabetes Research Center (ES-DRC), Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research (G.S.), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, International Translational Research and Medical Education (ITME) Consortium, "Federico II" University, Naples, Italy (J.G., D.S., G.S.); Department of Advanced Medical and Surgical Sciences (C.S., R.M., P. Ma., G.P.), and Department of Mental and Physical Health and Preventive Medicine (P.Ma.) University of Campania, Naples, Italy; Infectious Disease Unit, "Sant'Anna and San Sebastiano" Hospital, Caserta, Italy (V.M.)
| |
Collapse
|
38
|
Hou D, Zhang L, Hu Y, Yang G, Yu D. Bone Marrow Mesenchymal Stem Cell Exosomal miR-345-3p Ameliorates Cerebral Ischemia-reperfusion Injury by Targeting TRAF6. Curr Neurovasc Res 2023; 20:493-504. [PMID: 37670712 DOI: 10.2174/1567202620666230905121102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023]
Abstract
INTRODUCTION The purpose of this study was to investigate the effects of bone marrow mesenchymal stem cells (BMSCs) exosomal miR-345-3p and tumor necrosis factor receptorassociated factor 6 (TRAF6) on cerebral ischemia reperfusion (CIR) injury. Exosomes (Exos) derived from BMSCs were isolated and identified. PC12 (rat pheochromocytoma) cells were used to establish an oxygen and glucose deprivation/reoxygenation (OGD/R) model. METHODS Cell counting kit-8, TUNEL staining, lactate dehydrogenase staining, RT-qPCR, and western blotting were utilized for analyzing the functions of miR-345-3p about PC12 cells. Dualluciferase reporter experiment was then to confirm the link between miR-345-3p and TRAF6. Finally, using male SD rats, the middle cerebral artery occlusion (MCAO) model was constructed. Regulation of I/R damage in MCAO rats of miR-345-3p and TRAF6 were further explored in the changes of modified neurological severity score, cerebral infarction pictures, relative infarct volume, and histopathological changes. After OGD/R treatment, neuronal apoptosis was dramatically increased. After treatment with exosomal miR-345-3p, OGD/R-induced neuroapoptosis was dramatically inhibited. Exosomal miR-345-3p inhibited OGD/R-induced neuroapoptosis by downregulating the expression of TRAF6. However, the miR-345-3p inhibitor aggravated the changes caused by OGD/R. RESULTS The corresponding regulations of miR-345-3p were reversed with TRAF6 overexpression. The animal experiments in vivo further verified that miR-345-3p ameliorated brain I/R injury in MCAO rats by targeting TRAF6. CONCLUSION This study found that BMSCs-exosomal miR-345-3p protected against CIR injury by decreasing TRAF6.
Collapse
Affiliation(s)
- Dan Hou
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Lei Zhang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Yujie Hu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Guoshuai Yang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Dan Yu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| |
Collapse
|
39
|
Cui J, Li Y, Zhu M, Liu Y, Liu Y. Analysis of the Research Hotspot of Exosomes in Cardiovascular Disease: A Bibliometric-based Literature Review. Curr Vasc Pharmacol 2023; 21:316-345. [PMID: 37779407 DOI: 10.2174/0115701611249727230920042944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE To investigate the current status and development trend of research on exosomes in cardiovascular disease (CVD) using bibliometric analysis and to elucidate trending research topics. METHODS Research articles on exosomes in CVD published up to April 2022 were retrieved from the Web of Science database. Data were organized using Microsoft Office Excel 2019. CiteSpace 6.1 and VOSviewer 1.6.18 were used for bibliometric analysis and result visualization. RESULTS Overall, 256 original research publications containing 190 fundamental research publications and 66 clinical research publications were included. "Extracellular vesicle" was the most frequent research keyword, followed by "microrna," "apoptosis," and "angiogenesis." Most publications were from China (187, 73.05%), followed by the United States (57, 22.27%), the United Kingdom (7, 2.73%), and Japan (7, 2.73%). A systematic review of the publications revealed that myocardial infarction and stroke were the most popular topics and that exosomes and their contents, such as microRNAs (miRNAs), play positive roles in neuroprotection, inhibition of autophagy and apoptosis, promotion of angiogenesis, and protection of cardiomyocytes. CONCLUSION Research on exosomes in CVD has attracted considerable attention, with China having the most published studies. Fundamental research has focused on CVD pathogenesis; exosomes regulate the progression of CVD through biological processes, such as the inflammatory response, autophagy, and apoptosis. Clinical research has focused on biomarkers for CVD; studies on using miRNAs in exosomes as disease markers for diagnosis could become a future trend.
Collapse
Affiliation(s)
- Jing Cui
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiwen Li
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmeng Zhu
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Liu
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
40
|
Wang P, Dong S, Liu F, Liu A, Wang Z. MicroRNA-140-5p shuttled by microglia-derived extracellular vesicles attenuates subarachnoid hemorrhage-induced microglia activation and inflammatory response via MMD downregulation. Exp Neurol 2023; 359:114265. [PMID: 36336031 DOI: 10.1016/j.expneurol.2022.114265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND It is documented that microglia-secreted extracellular vesicles (microglia-EVs) exert neuroprotection which is important following subarachnoid hemorrhage (SAH). Herein, we focused on the mechanism of microglia-EVs harboring microRNA-140-5p (miR-140-5p) in SAH development. METHODS After the successful establishment of SAH rats, neurological function was evaluated, and behaviors were observed. Serum inflammatory factors (IL-1β and TNF-α) were quantified by ELISA, followed by the detection of microglial polarization by immunofluorescence. The relationship between miR-140-5p and monocyte to macrophage differentiation-associated (MMD) was evaluated using luciferase assay. Following the extraction of microglia and microglia-EVs, the transferring of miR-140-5p by microglia-EVs was assessed by co-culture experiments. SAH rats were treated with the EVs sourced from microglia overexpressing miR-140-5p (microglia-EVs-miR-140-5p) or EVs sourced from miR-140-5p-deficient microglia (microglia-EVs-miR-140-5p inhibitor) for in vivo effect assessment. RESULTS Microglia-EVs inhibited microglia activation and secretion of TNF-α and IL-1β by delivering miR-140-5p. Microglia-EVs could transmit miR-140-5p into microglia. Furthermore, microglia-EVs-miR-140-5p reduced the expression of its target MMD, resulting in blocked inflammatory response and activation of microglia in SAH rats by disrupting the PI3K/AKT and Erk1/2 signaling. CONCLUSION In summary, microglia-EVs transmitted miR-140-5p into microglia to downregulate MMD and finally contributed to neuroprotection in SAH rats.
Collapse
Affiliation(s)
- Pinyan Wang
- Department of Neurosurgery, the Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Siyuan Dong
- Department of Neurosurgery, the Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Fei Liu
- Department of Neurosurgery, the Third Xiangya Hospital of Central South University, Changsha 410013, PR China; Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, PR China.
| | - Aihua Liu
- Department of Neurosurgery, the Third Xiangya Hospital of Central South University, Changsha 410013, PR China; Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, PR China.
| | - Zhifei Wang
- Department of Neurosurgery, the Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| |
Collapse
|
41
|
Marangon D, Castro e Silva JH, Lecca D. Neuronal and Glial Communication via Non-Coding RNAs: Messages in Extracellular Vesicles. Int J Mol Sci 2022; 24:ijms24010470. [PMID: 36613914 PMCID: PMC9820657 DOI: 10.3390/ijms24010470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Extracellular vesicles (EVs) have been increasingly recognized as essential players in cell communication in many organs and systems, including the central nervous system (CNS). A proper interaction between neural cells is fundamental in the regulation of neurophysiological processes and its alteration could induce several pathological phenomena, such as neurodegeneration, neuroinflammation, and demyelination. EVs contain and transfer complex molecular cargoes typical of their cells of origin, such as proteins, lipids, carbohydrates, and metabolites to recipient cells. EVs are also enriched in non-coding RNAs (e.g., microRNAs, lncRNAs, and circRNA), which were formerly considered as cell-intrinsic regulators of CNS functions and pathologies, thus representing a new layer of regulation in the cell-to-cell communication. In this review, we summarize the most recent and advanced studies on the role of EV-derived ncRNAs in the CNS. First, we report the potential of neural stem cell-derived ncRNAs as new therapeutic tools for neurorepair. Then, we discuss the role of neuronal ncRNAs in regulating glia activation, and how alteration in glial ncRNAs influences neuronal survival and synaptic functions. We conclude that EV-derived ncRNAs can act as intercellular signals in the CNS to either propagate neuroinflammatory waves or promote reparative functions.
Collapse
|
42
|
Raza SHA, Wijayanti D, Pant SD, Abdelnour SA, Hashem NM, Amin A, Wani AK, Prakash A, Dawood MAO, Zan L. Exploring the physiological roles of circular RNAs in livestock animals. Res Vet Sci 2022; 152:726-735. [PMID: 36270182 DOI: 10.1016/j.rvsc.2022.09.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Circular RNAs (circRNAs) are a recently identified class of RNAs produced via back-splicing and covalent linkage between RNA ends, resulting in a circularized RNA molecule. Physiologically, circRNAs are known to influence a variety of biological pathways, and can also regulate transcription, post-transcription, RNA splicing, or interaction with other proteins or microRNAs (miRNAs). Functionally, circRNAs are known to competitively bind to various other RNA molecules including miRNAs and other competing endogenous RNA such as long noncoding RNA, thereby significantly influencing gene expression. Since gene expression is a crucial factor that underlies economically important livestock traits, it is likely that circRNAs significantly influence livestock traits like growth, milk production, reproduction, meat quality, hair follicle growth, and gametogenesis. Thousands of circRNAs have been recognized in different species of animals, and some of these circRNAs have also been shown to regulate stress responses that may be crucial for animal welfare. Therefore, in this review, we aim to highlight the biogenesis of circRNAs, along with its potential implications for livestock. The presented summary would offer a fundamental understanding of the molecular machinery that underlies circRNAs and associated biological phenomena and emphasize the need for further explorations into the role of circRNAs in the other productive, reproductive, and physiological attributes in animals.
Collapse
Affiliation(s)
- Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China; National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Dwi Wijayanti
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Sameer D Pant
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Sameh A Abdelnour
- Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Nesrein M Hashem
- Department of Animal and Fish Production, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria 21545, Egypt
| | - Ahmed Amin
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Atif Khurshid Wani
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University (144411), India
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, School of Medicine, USA
| | - Mahmoud A O Dawood
- Department of Animal Production, Faculty of Agriculture, Kafrelsheikh University, 33516 Egypt; Center for Applied Research on the Environment and Sustainability, The American University in Cairo, New Cairo 11835, Egypt
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China; National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
43
|
Jin M, Zhang S, Wang M, Li Q, Ren J, Luo Y, Sun X. Exosomes in pathogenesis, diagnosis, and therapy of ischemic stroke. Front Bioeng Biotechnol 2022; 10:980548. [PMID: 36588958 PMCID: PMC9800834 DOI: 10.3389/fbioe.2022.980548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Ischemic stroke is one of the major contributors to death and disability worldwide. Thus, there is an urgent need to develop early brain tissue perfusion therapies following acute stroke and to enhance functional recovery in stroke survivors. The morbidity, therapy, and recovery processes are highly orchestrated interactions involving the brain with other tissues. Exosomes are natural and ideal mediators of intercellular information transfer and recognized as biomarkers for disease diagnosis and prognosis. Changes in exosome contents express throughout the physiological process. Accumulating evidence demonstrates the use of exosomes in exploring unknown cellular and molecular mechanisms of intercellular communication and organ homeostasis and indicates their potential role in ischemic stroke. Inspired by the unique properties of exosomes, this review focuses on the communication, diagnosis, and therapeutic role of various derived exosomes, and their development and challenges for the treatment of cerebral ischemic stroke.
Collapse
Affiliation(s)
- Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Shuxia Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Mengchen Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Qiaoyu Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Jiahui Ren
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China,*Correspondence: Yun Luo, ; Xiaobo Sun,
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China,*Correspondence: Yun Luo, ; Xiaobo Sun,
| |
Collapse
|
44
|
Exosomes in Cerebral Ischemia-Reperfusion Injury: Current Perspectives and Future Challenges. Brain Sci 2022; 12:brainsci12121657. [PMID: 36552117 PMCID: PMC9776031 DOI: 10.3390/brainsci12121657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cerebral ischemia impedes the functional or metabolic demands of the central nervous system (CNS), which subsequently leads to irreversible brain damage. While recanalization of blocked vessels recovers cerebral blood flow, it can also aggravate brain injury, termed as ischemia/reperfusion (I/R) injury. Exosomes, nanometric membrane vesicles, attracted wide attention as carriers of biological macromolecules. In the brain, exosomes can be secreted by almost all types of cells, and their contents can be altered during the pathological and clinical processes of cerebral I/R injury. Herein, we will review the current literature on the possible role of cargos derived from exosomes and exosomes-mediated intercellular communication in cerebral I/R injury. The PubMed and Web of Science databases were searched through January 2015. The studies published in English were identified using search terms including "exosomes", "cerebral ischemia-reperfusion injury", "brain ischemia-reperfusion injury", and "stroke". We will also focus on the potential therapeutic effects of stem cell-derived exosomes and underlying mechanisms in cerebral I/R injury. Meanwhile, with the advantages of low immunogenicity and cytotoxicity, high bioavailability, and the capacity to pass through the blood-brain barrier, exosomes also attract more attention as therapeutic modalities for the treatment of cerebral I/R injury.
Collapse
|
45
|
Effects and Mechanisms of Exosomes from Different Sources in Cerebral Ischemia. Cells 2022; 11:cells11223623. [PMID: 36429051 PMCID: PMC9688936 DOI: 10.3390/cells11223623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Cerebral ischemia refers to the symptom of insufficient blood supply to the brain. Cells of many different origins participate in the process of repairing damage after cerebral ischemia occurs, in which exosomes secreted by the cells play important roles. For their characteristics, such as small molecular weight, low immunogenicity, and the easy penetration of the blood-brain barrier (BBB), exosomes can mediate cell-to-cell communication under pathophysiological conditions. In cerebral ischemia, exosomes can reduce neuronal damage and improve the brain microenvironment by regulating inflammation, mediating pyroptosis, promoting axonal growth, and stimulating vascular remodeling. Therefore, exosomes have an excellent application prospect for the treatment of cerebral ischemia. This article reviews the roles and mechanisms of exosomes from different sources in cerebral ischemia and provides new ideas for the prevention and treatment of cerebral ischemia.
Collapse
|
46
|
Chen Y, Zhu J, Ji J, Liu Z, Ren G. The role of microglial exosomes in brain injury. Front Cell Neurosci 2022; 16:1003809. [DOI: 10.3389/fncel.2022.1003809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Microglia are involved in immune responses to central nervous system (CNS) injury. Meanwhile, exosomes derived from microglia are important mediators of information and material exchange in brain, which play an important role in neuroprotective or damaging effects. Microglial exosomes contain a variety of molecular cargos, including microRNAs, soluble proteins, and lipids, which have regulatory effects on other types of cells and microenvironment in brain. In this review, we summarized microglial exosome characteristics, release patterns, pro-proliferative and pro-apoptotic effects on neurons and other glial cells, immunomodulatory effects, and regulation of the extracellular microenvironment. Understanding the relationship between microglia exosomes and brain injury can provide new targets for clinical treatment.
Collapse
|
47
|
Tan N, Xin W, Huang M, Mao Y. Mesenchymal stem cell therapy for ischemic stroke: Novel insight into the crosstalk with immune cells. Front Neurol 2022; 13:1048113. [PMID: 36425795 PMCID: PMC9679024 DOI: 10.3389/fneur.2022.1048113] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/17/2022] [Indexed: 09/29/2023] Open
Abstract
Stroke, a cerebrovascular accident, is prevalent and the second highest cause of death globally across patient populations; it is as a significant cause of morbidity and mortality. Mesenchymal stem cell (MSC) transplantation is emerging as a promising treatment for alleviating neurological deficits, as indicated by a great number of animal and clinical studies. The potential of regulating the immune system is currently being explored as a therapeutic target after ischemic stroke. This study will discuss recent evidence that MSCs can harness the immune system by interacting with immune cells to boost neurologic recovery effectively. Moreover, a notion will be given to MSCs participating in multiple pathological processes, such as increasing cell survival angiogenesis and suppressing cell apoptosis and autophagy in several phases of ischemic stroke, consequently promoting neurological function recovery. We will conclude the review by highlighting the clinical opportunities for MSCs by reviewing the safety, feasibility, and efficacy of MSCs therapy.
Collapse
Affiliation(s)
- Nana Tan
- Department of Health Management, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenqiang Xin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Huang
- Department of Health Management, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuling Mao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
48
|
Molecular Mechanism of the Protective Effects of M2 Microglia on Neurons: A Review Focused on Exosomes and Secretory Proteins. Neurochem Res 2022; 47:3556-3564. [PMID: 36222957 DOI: 10.1007/s11064-022-03760-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 10/17/2022]
Abstract
Microglia, as innate immune cells in the brain, closely monitor changes in the internal environment and participate in the maintenance of homeostasis in the central nervous system (CNS). Microglia can be polarized to the M1 or M2 phenotype in response to various stimuli in vivo or in vitro, affecting the functions of peripheral neurons. M2 microglia have attracted increasing attention in recent years owing to their beneficial effects on various diseases and injuries of the CNS, such as traumatic brain injury, stroke, Alzheimer's disease and multiple sclerosis. They exert neuroprotective effects by various mechanisms, e.g., suppressing inflammation, promoting the degradation of misfolded and aggregated proteins, promoting neurite growth, enhancing neurogenesis, inhibiting autophagy and apoptosis, promoting myelination, maintaining blood-brain barrier integrity, and enhancing phagocytic activity.This review summarizes the molecular mechanisms by which M2 microglia exert protective effects on neurons and provides a reference for the selection of therapeutic targets for CNS diseases.
Collapse
|
49
|
Yang F, Wang M, Guan X. Exosomes and mimics as novel delivery platform for cancer therapy. Front Pharmacol 2022; 13:1001417. [PMID: 36313380 PMCID: PMC9602403 DOI: 10.3389/fphar.2022.1001417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022] Open
Abstract
Exosomes are nano-sized biological extracellular vesicles transmitting information between cells and constituting a new intercellular communication mode. Exosomes have many advantages as an ideal drug delivery nanocarrier, including good biocompatibility, permeability, low toxicity, and low immunogenicity. Recently, exosomes have been used to deliver chemotherapeutic agents, natural drugs, nucleic acid drugs, and other antitumor drugs to treat many types of tumors. Due to the limited production of exosomes, synthetic exosome-mimics have been developed as an ideal platform for drug delivery. This review summarizes recent advances in the application of exosomes and exosome-mimics delivering therapeutic drugs in treating cancers.
Collapse
Affiliation(s)
- Fuxu Yang
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
- Key Laboratory of Pharmaceutics and Bioengineering, School of Medical Technology, Beihua University, Jilin, China
| | - Mingyue Wang
- Key Laboratory of Pharmaceutics and Bioengineering, School of Medical Technology, Beihua University, Jilin, China
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Xingang Guan
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
- *Correspondence: Xingang Guan,
| |
Collapse
|
50
|
Effects of miRNA-Modified Exosomes Alleviate Cerebral Ischemic Reperfusion Injury in Preclinical Studies: A Meta-Analysis. World Neurosurg 2022; 168:278-286.e2. [DOI: 10.1016/j.wneu.2022.09.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/05/2022]
|