1
|
Yang S, Guan Y, Zheng C, Xia X, Ma X, Jiang J. FOXO3-induced microRNA-128-3p promotes the progression of spinal cord injury in mice via regulating NLRP3 inflammasome-mediated pyroptosis. Front Immunol 2025; 16:1526721. [PMID: 40061945 PMCID: PMC11885150 DOI: 10.3389/fimmu.2025.1526721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/03/2025] [Indexed: 05/13/2025] Open
Abstract
Background Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate and complex pathophysiologic mechanisms. Pyroptosis, an inflammatory form of cell death triggered by certain inflammasomes, has a key role in a variety of inflammatory diseases, including SCI. However, it is unclear whether microRNAs (miRNAs), novel regulators in the SCI, are involved in SCI-induced pyroptosis. Methods Two GEO miRNA expression profiles (GSE158195 and GSE90452) were downloaded, and the differentially expressed miRNAs were analyzed by bioinformatics methods. An in vivo animal model and an in vitro cellular model of SCI were constructed in female C57BL/6 mice and BV-2 cells for studying the possible roles of FOXO3, miR-128-3p and NLRP3-mediated pyroptosis in SCI. Markers of ROS, cell pyroptosis and inflammation were measured by RT-qPCR, Western blotting, immunofluorescence, flow cytometry, and enzyme-linked immunosorbent assays. Histopathological changes in spinal cord tissue were detected using hematoxylin and eosin and immunohistochemical. The Basso-Beattie-Bresnahan (BBB) score was used to evaluate the motor function of mice in each group. Results Bioinformatics analysis of GSE158195 and GSE90452 datasets revealed a significant downregulation of miR-128-3p, a phenomenon that was consistently observed in the SCI mice model. Functionally, miR-128-3p upregulation improved functional behavioral recovery, relieved pathological injury, repressed oxidative stress, and alleviated pyroptosis and inflammation in the mouse SCI models. We also confirmed that Thioredoxin-interacting protein (TXNIP) was the target gene of miR-128-3p, and overexpression of TXNIP can effectively reverse the improvement of miR-128-3p in SCI cell model. Moreover, we found that transcription factor FOXO3 facilitated miR-128-3p expression, and its overexpression resulted in similar effects of miR-128-3p in the SCI cell model. Conclusion To the best of our knowledge, this is the first report demonstrating miR-128-3p improved secondary injury in SCI through the modulation of cell pyroptosis pathway. Our results suggest that FOXO3/miR-128-3p/TXNIP/NLRP3-mediated pyroptosis axis may be a potential therapeutic target for SCI.
Collapse
Affiliation(s)
| | | | - Chaojun Zheng
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | | | | |
Collapse
|
2
|
Moradi F, Mokhtari T. Role of NLRP3 Inflammasome in Chronic Pain and Alzheimer's Disease-A Review. J Biochem Mol Toxicol 2025; 39:e70071. [PMID: 39853846 PMCID: PMC11798427 DOI: 10.1002/jbt.70071] [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: 07/12/2024] [Revised: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 01/26/2025]
Abstract
The coexistence of Alzheimer's disease (AD) and chronic pain (CP) in the elderly population has been extensively documented, and a growing body of evidence supports the potential interconnections between these two conditions. This comprehensive review explores the mechanisms by which CP may contribute to the development and progression of AD, with a particular focus on neuroinflammatory pathways and the role of microglia, as well as the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. The review proposes that prolonged pain processing in critical brain regions can dysregulate the activity of the NLRP3 inflammasome within microglia, leading to the overproduction of pro-inflammatory cytokines and excessive oxidative stress in these regions. This aberrant microglial response also results in localized neuroinflammation in brain areas crucial for cognitive function. Additionally, CP as a persistent physiological and psychological stressor may be associated with hypothalamic-pituitary-adrenal (HPA) axis dysfunction, systemic inflammation, disruption of the blood-brain barrier (BBB), and neuroinflammation. These pathophysiological changes can cause morphological and functional impairments in brain regions responsible for cognition, memory, and neurotransmitter production, potentially contributing to the development and progression of CP-associated AD. Resultant neuroinflammation can further promote amyloid-beta (Aβ) plaque deposition, a hallmark of AD pathology. Potential therapeutic interventions targeting these neuroinflammatory pathways, particularly through the regulation of microglial NLRP3 activation, hold promise for improving outcomes in individuals with comorbid CP and AD. However, further research is required to fully elucidate the complex interplay between these conditions and develop effective treatment strategies.
Collapse
Affiliation(s)
- Fatemeh Moradi
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, 42 East Laurel Road, Stratford, NJ 08084, USA
| | - Tahmineh Mokhtari
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, People’s Republic of China
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, USA
| |
Collapse
|
3
|
Mohammadi S, Bashghareh A, Karimi-Zandi L, Mokhtari T. Understanding Role of Maternal Separation in Depression, Anxiety,and Pain Behaviour: A Mini Review of Preclinical Research With Focus on Neuroinflammatory Pathways. Int J Dev Neurosci 2025; 85:e70002. [PMID: 39895419 PMCID: PMC11838919 DOI: 10.1002/jdn.70002] [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: 10/14/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
Contact between mother and child is essential for the proper development of an infant's physiological systems, brain maturation and behavioural outcomes. Early life stress (ELS), which includes factors such as inadequate parental care and childhood abuse, significantly increases the risk of developing neuropsychiatric disorders, including anxiety and depression. This review examines the impact of maternal separation (MS) on depression, anxiety and pain behaviour, with a particular emphasis on neuroinflammatory pathways. Experiences of ELS can adversely affect the maturation of neurotransmitter systems and associated neural circuits that are crucial for processing painful stimuli and regulating anxiety and depression. Stressful experiences trigger inflammatory processes in the brain, initiating immune responses in neural cells and stimulating the production of pro-inflammatory cytokines. In mammals, MS serves as a significant stressor that activates the hypothalamic-pituitary-adrenal (HPA) axis and other stress-related systems, leading to increased immune challenges and heightened pain sensitivity in adulthood due to systemic inflammation. Key inflammatory mediators, such as IL-1β, IL-6 and TNF-α, play critical roles in the development of pathological pain, while the activation of microglia releases inflammatory mediators that contribute to neurological dysfunction and the pathophysiology of stress, depression and anxiety. Moreover, therapeutics targeting oxidative stress and inflammation have shown promise in alleviating affective disorders following MS. This review discusses potential pathways, with a primary focus on neuroinflammatory mechanisms and the therapeutic strategies that may mitigate the adverse effects of MS. There is a pressing need for further research to elucidate the underlying pathways and identify effective interventions to improve mental health outcomes in individuals affected by MS.
Collapse
Affiliation(s)
- Shima Mohammadi
- Department of Neuroscience, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Alieh Bashghareh
- Department of Anatomy, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Leila Karimi-Zandi
- School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Tahmineh Mokhtari
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA 95616, United States
| |
Collapse
|
4
|
Wang JL, Huang QM, Hu DX, Zhang WJ. Therapeutic effect of exosomes derived from Schwann cells in the repair of peripheral nerve injury. Life Sci 2024; 357:123086. [PMID: 39357794 DOI: 10.1016/j.lfs.2024.123086] [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: 07/18/2024] [Revised: 09/22/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Peripheral nerve injury (PNI) can cause nerve demyelination, neuronal apoptosis, axonal atrophy, inflammatory infiltration, glial scar formation, and other pathologies that can lead to sensory and motor dysfunction and seriously affect the psychosomatic health of patients. There is currently no effective treatment method, so exploring a promising treatment method is of great significance. Several studies have revealed the therapeutic roles of Schwann cells (SCs) and their exosomes in nerve injury repair. Exosomes are extracellular nanovesicles secreted by cells that act as key molecules in intercellular communication. Progress has been made in understanding the role of exosomes derived from SCs (SC-EXOs) in peripheral nerve regeneration, including the promotion of axonal regeneration and myelin formation, anti-inflammation, vascular regeneration, neuroprotection, and neuroregulation. Therefore, in this paper, we summarize the functional characteristics of SC-EXOs and discuss their potential therapeutic effects on PNI repair as well as some existing problems and future challenges.
Collapse
Affiliation(s)
- Jia-Ling Wang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Qi-Ming Huang
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
5
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-w] [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: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
6
|
Ebrahimi B, Mokhtari T, Ghaffari N, Adabi M, Hassanzadeh G. Acellular spinal cord scaffold containing quercetin-encapsulated nanoparticles plays an anti-inflammatory role in functional recovery from spinal cord injury in rats. Inflammopharmacology 2024; 32:2505-2524. [PMID: 38702577 DOI: 10.1007/s10787-024-01478-z] [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: 01/23/2024] [Accepted: 04/09/2024] [Indexed: 05/06/2024]
Abstract
Inflammatory responses play a crucial role in the pathophysiology of spinal cord injury (SCI) and developing new approaches to establish an anti-inflammatory environment for the promotion of neuroregeneration holds promise as a potential approach. In this study, our aim was to investigate the potential of combining an acellular spinal cord scaffold (ASCS) with quercetin-loaded bovine serum albumin (Qu/BSA) nanoparticles (NPs) for the treatment of SCI. The ASCS was prepared using physical and chemical methods, while the Qu/BSA NPs were prepared through a desolvation technique. The NPs exhibited favorable characteristics, including a mean size of 203 nm, a zeta potential of -38, and an encapsulation efficiency of 96%. Microscopic evaluation confirmed the successful distribution of NPs on the walls of ASCS. Animal studies revealed that Qu/BSA NPs group exhibited a significant decrease in NLRP3, ASC, and Casp1 gene expression compared to the SCI group (p < 0.0001). The findings indicated a significant decrease in the NLRP3, ASC, and Casp1 protein level between the Qu/BSA/ASCS group and the SCI group (p < 0.0001). Moreover, treatment with ASCS containing either blank BSA (B/BSA) NPs or Qu/BSA NPs effectively promoted functional recovery via increasing the amount of nestin- and glial fibrillary acidic protein (GFAP)-positive cells in the site of injury. Notably, Qu/BSA/ASCS exhibited superior outcomes compared to B/BSA/ASCS. Overall, the combination of ASCS with the Qu delivery system presents a promising therapeutic approach for SCI by inhibiting inflammatory responses and promoting neuroregeneration, leading to the restoration of motor function in animals. This study demonstrates the potential of utilizing biomaterials and NPs to enhance the effectiveness of SCI treatment.
Collapse
Affiliation(s)
- Babak Ebrahimi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Mokhtari
- Department of Pharmacology, Hubei University of Medicine, Shiyan, China.
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.
| | - Neda Ghaffari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Neurosciences and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Si Y, Hayat MA, Hu J. NSPCs-ES: mechanisms and functional impact on central nervous system diseases. Biomed Mater 2024; 19:042011. [PMID: 38916246 DOI: 10.1088/1748-605x/ad5819] [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: 01/04/2024] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
Patients with central neuronal damage may suffer severe consequences, but effective therapies remain unclear. Previous research has established the transplantation of neural stem cells that generate new neurons to replace damaged ones. In a new field of scientific research, the extracellular secretion of NPSCs (NSPCs-ES) has been identified as an alternative to current chemical drugs. Many preclinical studies have shown that NSPCs-ES are effective in models of various central nervous system diseases (CNS) injuries, from maintaining functional structures at the cellular level to providing anti-inflammatory functions at the molecular level, as well as improving memory and motor functions, reducing apoptosis in neurons, and mediating multiple signaling pathways. The NSPC-ES can travel to the damaged tissue and exert a broad range of therapeutic effects by supporting and nourishing damaged neurons. However, gene editing and cell engineering techniques have recently improved therapeutic efficacy by modifying NSPCs-ES. Consequently, future research and application of NSPCs-ES may provide a novel strategy for the treatment of CNS diseases in the future. In this review, we summarize the current progress on these aspects.
Collapse
Affiliation(s)
- Yu Si
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, People's Republic of China
| | - Muhammad Abid Hayat
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, People's Republic of China
| | - Jiabo Hu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, People's Republic of China
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, People's Republic of China
- Zhenjiang Blood Center, Zhenjiang, Jiangsu 212013, People's Republic of China
| |
Collapse
|
8
|
Ghaffari N, Mokhtari T, Adabi M, Ebrahimi B, Kamali M, Gholaminejhad M, Hassanzadeh G. Neurological recovery and neurogenesis by curcumin sustained-release system cross-linked with an acellular spinal cord scaffold in rat spinal cord injury: Targeting NLRP3 inflammasome pathway. Phytother Res 2024; 38:2669-2686. [PMID: 38500263 DOI: 10.1002/ptr.8179] [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: 07/01/2023] [Revised: 01/03/2024] [Accepted: 02/11/2024] [Indexed: 03/20/2024]
Abstract
In the context of treating spinal cord injury (SCI), the modulation of inflammatory responses, and the creation of a suitable region for tissue regeneration may present a promising approach. This study aimed to evaluate the effects of curcumin (Cur)-loaded bovine serum albumin nanoparticles (Cur-BSA NPs) cross-linked with an acellular spinal cord scaffold (ASCS) on the functional recovery in a rat model of SCI. We developed an ASCS using chemical and physical methods. Cur-BSA, and blank (B-BSA) NPs were fabricated and cross-linked with ASCS via EDC-NHS, resulting in the production of Cur-ASCS and B-ASCS. We assessed the properties of scaffolds and NPs as well as their cross-links. Finally, using a male rat hemisection model of SCI, we investigated the consequences of the resulting scaffolds. The inflammatory markers, neuroregeneration, and functional recovery were evaluated. Our results showed that Cur was efficiently entrapped at the rate of 42% ± 1.3 in the NPs. Compared to B-ASCS, Cur-ASCS showed greater effectiveness in the promotion of motor recovery. The implantation of both scaffolds could increase the migration of neural stem cells (Nestin- and GFAP-positive cells) following SCI with the superiority of Cur-ASCS. Cur-ASCS was successful to regulate the gene expression and protein levels of NLRP3, ASC, and Casp1in the spinal cord lesion. Our results indicate that using ASCS can lead to the entrance of cells into the scaffold and promote neurogenesis. However, Cur-ASCS had greater effects in terms of inflammation relief and enhanced neurogenesis.
Collapse
Affiliation(s)
- Neda Ghaffari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Ebrahimi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Kamali
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Gholaminejhad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurosciences and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Mokhtari T, Irandoost E, Sheikhbahaei F. Stress, pain, anxiety, and depression in endometriosis-Targeting glial activation and inflammation. Int Immunopharmacol 2024; 132:111942. [PMID: 38565045 DOI: 10.1016/j.intimp.2024.111942] [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: 02/10/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
Endometriosis (EM) is a gynecological inflammatory disease often accompanied by stress, chronic pelvic pain (CPP), anxiety, and depression, leading to a diminished quality of life. This review aims to discuss the relationship between systemic and local inflammatory responses in the central nervous system (CNS), focusing on glial dysfunctions (astrocytes and microglia) as in critical brain regions involved in emotion, cognition, pain processing, anxiety, and depression. The review presents that EM is connected to increased levels of pro-inflammatory cytokines in the circulation. Additionally, chronic stress and CPP as stressors may contribute to the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, depleting the production of inflammatory mediators in the circulatory system and the brain. The systemic cytokines cause blood-brain barrier (BBB) breakdown, activate microglia in the brain, and lead to neuroinflammation. Furthermore, CPP may induce neuronal morphological alterations in critical regions through central sensitization and the activation of glial cells. The activation of glial cells, particularly the polarization of microglia, leads to the activation of the NLRP3 inflammasome and the overproduction of inflammatory cytokines. These inflammatory cytokines interact with the signaling pathways involved in neural plasticity. Additionally, persistent inflammatory conditions in the brain lead to neuronal death, which is correlated with a reduced volume of key brain regions such as the hippocampus. This review highlights the involvement of glial cells in the pathogenesis of the mental comorbidities of EM (i.e., pain, anxiety, and depression) and to discuss potential therapeutic approaches for targeting the inflammation and activation of microglia in key brain regions.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- Department of Pharmacology, Hubei University of Medicine, Shiyan, China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Elnaz Irandoost
- Sarem Women's Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Fatemeh Sheikhbahaei
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
10
|
Sintakova K, Romanyuk N. The role of small extracellular vesicles and microRNA as their cargo in the spinal cord injury pathophysiology and therapy. Front Neurosci 2024; 18:1400413. [PMID: 38774785 PMCID: PMC11106386 DOI: 10.3389/fnins.2024.1400413] [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: 03/13/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with a complex pathology that affects a significant portion of the population and causes long-term consequences. After primary injury, an inflammatory cascade of secondary injury occurs, followed by neuronal cell death and glial scar formation. Together with the limited regenerative capacity of the central nervous system, these are the main reasons for the poor prognosis after SCI. Despite recent advances, there is still no effective treatment. Promising therapeutic approaches include stem cells transplantation, which has demonstrated neuroprotective and immunomodulatory effects in SCI. This positive effect is thought to be mediated by small extracellular vesicles (sEVs); membrane-bound nanovesicles involved in intercellular communication through transport of functional proteins and RNA molecules. In this review, we summarize the current knowledge about sEVs and microRNA as their cargo as one of the most promising therapeutic approaches for the treatment of SCI. We provide a comprehensive overview of their role in SCI pathophysiology, neuroprotective potential and therapeutic effect.
Collapse
Affiliation(s)
- Kristyna Sintakova
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czechia
| | - Nataliya Romanyuk
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
11
|
Nie X, Yuan T, Yu T, Yun Z, Yu T, Liu Q. Non-stem cell-derived exosomes: a novel therapeutics for neurotrauma. J Nanobiotechnology 2024; 22:108. [PMID: 38475766 DOI: 10.1186/s12951-024-02380-0] [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: 10/23/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Neurotrauma, encompassing traumatic brain injuries (TBI) and spinal cord injuries (SCI) impacts a significant portion of the global population. While spontaneous recovery post-TBI or SCI is possible, recent advancements in cell-based therapies aim to bolster these natural reparative mechanisms. Emerging research indicates that the beneficial outcomes of such therapies might be largely mediated by exosomes secreted from the administered cells. While stem cells have garnered much attention, exosomes derived from non-stem cells, including neurons, Schwann cells, microglia, and vascular endothelial cells, have shown notable therapeutic potential. These exosomes contribute to angiogenesis, neurogenesis, and axon remodeling, and display anti-inflammatory properties, marking them as promising agents for neurorestorative treatments. This review provides an in-depth exploration of the current methodologies, challenges, and future directions regarding the therapeutic role of non-stem cell-derived exosomes in neurotrauma.
Collapse
Affiliation(s)
- Xinyu Nie
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Tianyang Yuan
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Tong Yu
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Zhihe Yun
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Tao Yu
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Qinyi Liu
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China.
| |
Collapse
|
12
|
Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain 2024; 147:766-793. [PMID: 37975820 DOI: 10.1093/brain/awad392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a leading cause of lifelong disabilities. Permanent sensory, motor and autonomic impairments after SCI are substantially attributed to degeneration of spinal cord neurons and axons, and disintegration of neural network. To date, minimal regenerative treatments are available for SCI with an unmet need for new therapies to reconstruct the damaged spinal cord neuron-glia network and restore connectivity with the supraspinal pathways. Multipotent neural precursor cells (NPCs) have a unique capacity to generate neurons, oligodendrocytes and astrocytes. Due to this capacity, NPCs have been an attractive cell source for cellular therapies for SCI. Transplantation of NPCs has been extensively tested in preclinical models of SCI in the past two decades. These studies have identified opportunities and challenges associated with NPC therapies. While NPCs have the potential to promote neuroregeneration through various mechanisms, their low long-term survival and integration within the host injured spinal cord limit the functional benefits of NPC-based therapies for SCI. To address this challenge, combinatorial strategies have been developed to optimize the outcomes of NPC therapies by enriching SCI microenvironment through biomaterials, genetic and pharmacological therapies. In this review, we will provide an in-depth discussion on recent advances in preclinical NPC-based therapies for SCI. We will discuss modes of actions and mechanism by which engrafted NPCs contribute to the repair process and functional recovery. We will also provide an update on current clinical trials and new technologies that have facilitated preparation of medical-grade human NPCs suitable for transplantation in clinical studies.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
| | - Ben Borys
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
13
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [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: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
14
|
Mokhtari T, Uludag K. Role of NLRP3 Inflammasome in Post-Spinal-Cord-Injury Anxiety and Depression: Molecular Mechanisms and Therapeutic Implications. ACS Chem Neurosci 2024; 15:56-70. [PMID: 38109051 DOI: 10.1021/acschemneuro.3c00596] [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: 12/19/2023] Open
Abstract
The majority of research on the long-term effects of spinal cord injury (SCI) has primarily focused on neuropathic pain (NP), psychological issues, and sensorimotor impairments. Among SCI patients, mood disorders, such as anxiety and depression, have been extensively studied. It has been found that chronic stress and NP have negative consequences and reduce the quality of life for individuals living with SCI. Our review examined both human and experimental evidence to explore the connection between mood changes following SCI and inflammatory pathways, with a specific focus on NLRP3 inflammasome signaling. We observed increased proinflammatory factors in the blood, as well as in the brain and spinal cord tissues of SCI models. The NLRP3 inflammasome plays a crucial role in various diseases by controlling the release of proinflammatory molecules like interleukin 1β (IL-1β) and IL-18. Dysregulation of the NLRP3 inflammasome in key brain regions associated with pain processing, such as the prefrontal cortex and hippocampus, contributes to the development of mood disorders following SCI. In this review, we summarized recent research on the expression and regulation of components related to NLRP3 inflammasome signaling in mood disorders following SCI. Finally, we discussed potential therapeutic approaches that target the NLRP3 inflammasome and regulate proinflammatory cytokines as a way to treat mood disorders following SCI.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Kadir Uludag
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, People's Republic of China
| |
Collapse
|
15
|
Qin B, Hu XM, Huang YX, Yang RH, Xiong K. A New Paradigm in Spinal Cord Injury Therapy: from Cell-free Treatment to Engineering Modifications. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:656-673. [PMID: 37076458 DOI: 10.2174/1871527322666230418090857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/20/2023] [Accepted: 02/06/2023] [Indexed: 04/21/2023]
Abstract
Spinal cord injury (SCI) is an intractable and poorly prognostic neurological disease, and current treatments are still unable to cure it completely and avoid sequelae. Extracellular vesicles (EVs), as important carriers of intercellular communication and pharmacological effects, are considered to be the most promising candidates for SCI therapy because of their low toxicity and immunogenicity, their ability to encapsulate endogenous bioactive molecules (e.g., proteins, lipids, and nucleic acids), and their ability to cross the blood-brain/cerebrospinal barriers. However, poor targeting, low retention rate, and limited therapeutic efficacy of natural EVs have bottlenecked EVs-based SCI therapy. A new paradigm for SCI treatment will be provided by engineering modified EVs. Furthermore, our limited understanding of the role of EVs in SCI pathology hinders the rational design of novel EVbased therapeutic approaches. In this study, we review the pathophysiology after SCI, especially the multicellular EVs-mediated crosstalk; briefly describe the shift from cellular to cell-free therapies for SCI treatment; discuss and analyze the issues related to the route and dose of EVs administration; summarize and present the common strategies for EVs drug loading in the treatment of SCI and point out the shortcomings of these drug loading methods; finally, we analyze and highlight the feasibility and advantages of bio-scaffold-encapsulated EVs for SCI treatment, providing scalable insights into cell-free therapy for SCI.
Collapse
Affiliation(s)
- Bo Qin
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China
| | - Xi-Min Hu
- Clinical Medicine Eight-year Program, 02 Class, 17 Grade, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yan-Xia Huang
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Rong-Hua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
- Hunan Key Laboratory of Ophthalmology, Changsha, 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| |
Collapse
|
16
|
Chamanara S, Hozouri V, Irandoost E. Inhibition of NLRP3 inflammasome-A potential mechanistic therapeutic for treatment of polycystic ovary syndrome? J Biochem Mol Toxicol 2024; 38:e23592. [PMID: 38054794 DOI: 10.1002/jbt.23592] [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: 05/02/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023]
Abstract
This review article explores the relationship between the NOD-like receptor protein 3 (NLRP3) inflammasome and the risk of developing polycystic ovary syndrome (PCOS). The NLRP3 inflammasome, a fundamental element of the innate immune system, plays a crucial role in the production of proinflammatory mediators and pyroptosis, a type inflammatory cell death. We conducted a thorough search on scientific databases to gather relevant information on this topic, utilizing relevant keywords. The reviewed studies indicated a correlation between PCOS and a higher incidence of granulosa cell (GC) death and the presence of ovarian tissue fibrosis. NLRP3 inflammasome stimulation and subsequent pyroptosis in GCs play a significant role in the pathophysiology of PCOS. Active NLRP3 inflammasome is involved in the production of inflammatory mediators like interleukin-1β (IL-1β) and IL-18, contributing to the development of PCOS, particularly in overweight patients. Therefore, inhibiting NLRP3 activation and pyroptosis could potentially offer novel therapeutic strategies for PCOS. Some limited studies have explored the use of agents with antioxidant and anti-inflammatory properties, as well as gene therapy approaches, to target the NLRP3 and pyroptosis signaling pathways. This study overview the understanding of the relationship between NLRP3 inflammasome activation, pyroptosis, and PCOS. It highlights the potential of targeting the NLRP3 inflammasome as an approach for treating PCOS. Nonetheless, further research and clinical trials are imperative to validate these results and explore the effectiveness of NLRP3 inflammasome inhibition in the management of PCOS.
Collapse
Affiliation(s)
- Solmaz Chamanara
- Department of Gynecology and Obstetrics, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Vahid Hozouri
- Internal Medicine Department, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Elnaz Irandoost
- Department of Gynecology and Obstetrics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Nazerian Y, Nazerian A, Mohamadi-Jahani F, Sodeifi P, Jafarian M, Javadi SAH. Hydrogel-encapsulated extracellular vesicles for the regeneration of spinal cord injury. Front Neurosci 2023; 17:1309172. [PMID: 38156267 PMCID: PMC10752990 DOI: 10.3389/fnins.2023.1309172] [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: 10/07/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
Spinal cord injury (SCI) is a critical neurological condition that may impair motor, sensory, and autonomous functions. At the cellular level, inflammation, impairment of axonal regeneration, and neuronal death are responsible for SCI-related complications. Regarding the high mortality and morbidity rates associated with SCI, there is a need for effective treatment. Despite advances in SCI repair, an optimal treatment for complete recovery after SCI has not been found so far. Therefore, an effective strategy is needed to promote neuronal regeneration and repair after SCI. In recent years, regenerative treatments have become a potential option for achieving improved functional recovery after SCI by promoting the growth of new neurons, protecting surviving neurons, and preventing additional damage to the spinal cord. Transplantation of cells and cells-derived extracellular vesicles (EVs) can be effective for SCI recovery. However, there are some limitations and challenges related to cell-based strategies. Ethical concerns and limited efficacy due to the low survival rate, immune rejection, and tumor formation are limitations of cell-based therapies. Using EVs is a helpful strategy to overcome these limitations. It should be considered that short half-life, poor accumulation, rapid clearance, and difficulty in targeting specific tissues are limitations of EVs-based therapies. Hydrogel-encapsulated exosomes have overcome these limitations by enhancing the efficacy of exosomes through maintaining their bioactivity, protecting EVs from rapid clearance, and facilitating the sustained release of EVs at the target site. These hydrogel-encapsulated EVs can promote neuroregeneration through improving functional recovery, reducing inflammation, and enhancing neuronal regeneration after SCI. This review aims to provide an overview of the current research status, challenges, and future clinical opportunities of hydrogel-encapsulated EVs in the treatment of SCI.
Collapse
Affiliation(s)
- Yasaman Nazerian
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Fereshteh Mohamadi-Jahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parastoo Sodeifi
- School of Medicine, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Maryam Jafarian
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Hossein Javadi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurosurgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Xu X, Li L, Wang B, Shi B. Caffeic acid phenethyl ester ameliorates titanium particle-induced bone loss and inflammatory reaction in a mouse acute model. Biochem Biophys Res Commun 2023; 681:47-54. [PMID: 37751634 DOI: 10.1016/j.bbrc.2023.09.049] [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: 07/17/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 09/28/2023]
Abstract
With the increasing clinical application of dental and orthopedic implants, the problem of peri-implant osteolysis has attracted attention. The inflammatory response and osteoclast differentiation induced by wear particles play an important role in peri-implant bone loss. However, the treatment of peri-implant osteolysis is still lacking. In the present study, we investigated the effect of caffeic acid phenethyl ester (CAPE) on titanium particles induced bone loss in a mouse model. We found that CAPE significantly suppressed titanium particle-induced bone loss in vivo. CAPE treatment decreased ratio of nuclear factor kappa B receptor activator ligand (RANKL)/osteoprotegerin (OPG) and subsequently reduced osteoclastogenesis in the mouse model. In addition, CAPE downregulated the expression and secretion of interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α) stimulated by titanium particles in vivo. In summary, we conclude that CAPE prevent the titanium particles-induced bone loss.
Collapse
Affiliation(s)
- Xiaoqian Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Lei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Beike Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bin Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
19
|
Xuan L, Hu Z, Jiang Z, Zhang C, Sun X, Ming W, Liu H, Qiao R, Shen L, Liu S, Wang G, Wen L, Luan Z, Yin J. Pregnane X receptor (PXR) deficiency protects against spinal cord injury by activating NRF2/HO-1 pathway. CNS Neurosci Ther 2023; 29:3460-3478. [PMID: 37269088 PMCID: PMC10580351 DOI: 10.1111/cns.14279] [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: 10/28/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023] Open
Abstract
INTRODUCTION As a devastating neurological disease, spinal cord injury (SCI) results in severe tissue loss and neurological dysfunction. Pregnane X receptor (PXR) is a ligand-activated nuclear receptor with a major regulatory role in xenobiotic and endobiotic metabolism and recently has been implicated in the central nervous system. In the present study, we aimed to investigate the role and mechanism of PXR in SCI. METHODS The clip-compressive SCI model was performed in male wild-type C57BL/6 (PXR+/+ ) and PXR-knockout (PXR-/- ) mice. The N2a H2 O2 -induced injury model mimicked the pathological process of SCI in vitro. Pregnenolone 16α-carbonitrile (PCN), a mouse-specific PXR agonist, was used to activate PXR in vivo and in vitro. The siRNA was applied to knock down the PXR expression in vitro. Transcriptome sequencing analysis was performed to discover the relevant mechanism, and the NRF2 inhibitor ML385 was used to validate the involvement of PXR in influencing the NRF2/HO-1 pathway in the SCI process. RESULTS The expression of PXR decreased after SCI and reached a minimum on the third day. In vivo, PXR knockout significantly improved the motor function of mice after SCI, meanwhile, inhibited apoptosis, inflammation, and oxidative stress induced by SCI. On the contrary, activation of PXR by PCN negatively influenced the recovery of SCI. Mechanistically, transcriptome sequencing analysis revealed that PXR activation downregulated the mRNA level of heme oxygenase-1 (HO-1) after SCI. We further verified that PXR deficiency activated the NRF2/HO-1 pathway and PXR activation inhibited this pathway in vitro. CONCLUSION PXR is involved in the recovery of motor function after SCI by regulating NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- Li‐Na Xuan
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhen‐Xin Hu
- Department of OrthopedicsThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhen‐Fu Jiang
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Cong Zhang
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Xiao‐Wan Sun
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Wen‐Hua Ming
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Hui‐Tao Liu
- Department of OrthopedicsTaizhou Hospital of Zhejiang ProvinceLinhaiChina
| | - Rong‐Fang Qiao
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Lin‐Jie Shen
- Department of GastroenterologyNingbo First HospitalNingboChina
| | - Shao‐Bo Liu
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Guan‐Yu Wang
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Lin Wen
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Zhi‐Lin Luan
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic DiseasesDalianChina
| | - Jian Yin
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
20
|
Nazari S, Pourmand SM, Motevaseli E, Hassanzadeh G. Mesenchymal stem cells (MSCs) and MSC-derived exosomes in animal models of central nervous system diseases: Targeting the NLRP3 inflammasome. IUBMB Life 2023; 75:794-810. [PMID: 37278718 DOI: 10.1002/iub.2759] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023]
Abstract
The NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome is a multimeric protein complex that is engaged in the innate immune system and plays a vital role in inflammatory reactions. Activation of the NLRP3 inflammasome and subsequent release of proinflammatory cytokines can be triggered by microbial infection or cellular injury. The NLRP3 inflammasome has been implicated in the pathogenesis of many disorders affecting the central nervous system (CNS), ranging from stroke, traumatic brain injury, and spinal cord injury to Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, and depression. Furthermore, emerging evidence has suggested that mesenchymal stem cells (MSCs) and their exosomes may modulate NLRP3 inflammasome activation in a way that might be promising for the therapeutic management of CNS diseases. In the present review, particular focus is placed on highlighting and discussing recent scientific evidence regarding the regulatory effects of MSC-based therapies on the NLRP3 inflammasome activation and their potential to counteract proinflammatory responses and pyroptotic cell death in the CNS, thereby achieving neuroprotective impacts and improvement in behavioral impairments.
Collapse
Affiliation(s)
- Shahrzad Nazari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Pourmand
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Li Y, Fang B. Neural stem cell-derived extracellular vesicles: The light of central nervous system diseases. Biomed Pharmacother 2023; 165:115092. [PMID: 37406512 DOI: 10.1016/j.biopha.2023.115092] [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: 04/16/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023] Open
Abstract
Central nervous system (CNS) diseases are the leading cause of death worldwide. By performing compensatory functions and improving the inflammatory microenvironment, the transplantation of neural stem cells (NSCs) can promote functional recovery from brain injury, aging, brain tumours, and other diseases. However, the ability of NSCs to differentiate into neurons is limited, and they are associated with a risk of tumourigenicity. NSC-derived extracellular vesicles (NSC-EVs) can modulate the local microenvironment of the nervous system as well as distant neuronal functions. Thus, cell-free therapy may be a novel remedy for CNS disorders. This article reviews the characteristics, contents, and mechanisms of action of NSC-EVs as well as their roles and application prospects in various CNS diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
22
|
Yue W, Shen J. Local Delivery Strategies for Peptides and Proteins into the CNS: Status Quo, Challenges, and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:810. [PMID: 37375758 DOI: 10.3390/ph16060810] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Over the past decades, peptides and proteins have been increasingly important in the treatment of various human diseases and conditions owing to their specificity, potency, and minimized off-target toxicity. However, the existence of the practically impermeable blood brain barrier (BBB) limits the entry of macromolecular therapeutics into the central nervous systems (CNS). Consequently, clinical translation of peptide/protein therapeutics for the treatment of CNS diseases has been limited. Over the past decades, developing effective delivery strategies for peptides and proteins has gained extensive attention, in particular with localized delivery strategies, due to the fact that they are capable of circumventing the physiological barrier to directly introduce macromolecular therapeutics into the CNS to improve therapeutic effects and reduce systemic side effects. Here, we discuss various local administration and formulation strategies that have shown successes in the treatment of CNS diseases using peptide/protein therapeutics. Lastly, we discuss challenges and future perspectives of these approaches.
Collapse
Affiliation(s)
- Weizhou Yue
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Jie Shen
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
23
|
Patel S, Guo MK, Abdul Samad M, Howe KL. Extracellular vesicles as biomarkers and modulators of atherosclerosis pathogenesis. Front Cardiovasc Med 2023; 10:1202187. [PMID: 37304965 PMCID: PMC10250645 DOI: 10.3389/fcvm.2023.1202187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 06/13/2023] Open
Abstract
Extracellular vesicles (EVs) are small, lipid bilayer-enclosed structures released by various cell types that play a critical role in intercellular communication. In atherosclerosis, EVs have been implicated in multiple pathophysiological processes, including endothelial dysfunction, inflammation, and thrombosis. This review provides an up-to-date overview of our current understanding of the roles of EVs in atherosclerosis, emphasizing their potential as diagnostic biomarkers and their roles in disease pathogenesis. We discuss the different types of EVs involved in atherosclerosis, the diverse cargoes they carry, their mechanisms of action, and the various methods employed for their isolation and analysis. Moreover, we underscore the importance of using relevant animal models and human samples to elucidate the role of EVs in disease pathogenesis. Overall, this review consolidates our current knowledge of EVs in atherosclerosis and highlights their potential as promising targets for disease diagnosis and therapy.
Collapse
Affiliation(s)
- Sarvatit Patel
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Mandy Kunze Guo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Majed Abdul Samad
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
24
|
Li X, Zhu Y, Wang Y, Xia X, Zheng JC. Neural stem/progenitor cell-derived extracellular vesicles: A novel therapy for neurological diseases and beyond. MedComm (Beijing) 2023; 4:e214. [PMID: 36776763 PMCID: PMC9905070 DOI: 10.1002/mco2.214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
As bilayer lipid membrane vesicles secreted by neural stem/progenitor cells (NSCs), NSC-derived extracellular vesicles (NSC-EVs) have attracted growing attention for their promising potential to serve as novel therapeutic agents in treatment of neurological diseases due to their unique physicochemical characteristics and biological functions. NSC-EVs exhibit advantages such as stable physical and chemical properties, low immunogenicity, and high penetration capacity to cross blood-brain barrier to avoid predicaments of the clinical applications of NSCs that include autoimmune responses, ethical/religious concerns, and the problematic logistics of acquiring fetal tissues. More importantly, NSC-EVs inherit excellent neuroprotective and neuroregenerative potential and immunomodulatory capabilities from parent cells, and display outstanding therapeutic effects on mitigating behavioral alterations and pathological phenotypes of patients or animals with neurological diseases. In this review, we first comprehensively summarize the progress in functional research and application of NSC-EVs in different neurological diseases, including neurodegenerative diseases, acute neurological diseases, dementia/cognitive dysfunction, and peripheral diseases. Next, we provide our thoughts on current limitations/concerns as well as tremendous potential of NSC-EVs in clinical applications. Last, we discuss future directions of further investigations on NSC-EVs and their probable applications in both basic and clinical research.
Collapse
Affiliation(s)
- Xiangyu Li
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
| | - Yingbo Zhu
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative TherapyYangzhi Rehabilitation Hospital, Tongji UniversityShanghaiChina
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Frontiers Science Center of Nanocatalytic MedicineTongji University School of MedicineShanghaiChina
- Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, Tongji University School of MedicineShanghaiChina
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Tongji UniversityMinistry of EducationShanghaiChina
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Frontiers Science Center of Nanocatalytic MedicineTongji University School of MedicineShanghaiChina
- Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, Tongji University School of MedicineShanghaiChina
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Tongji UniversityMinistry of EducationShanghaiChina
| |
Collapse
|
25
|
Yin T, Li Y, Sung P, Chiang JY, Shao P, Yip H, Lee MS. Adipose-derived mesenchymal stem cells overexpressing prion improve outcomes via the NLRP3 inflammasome/DAMP signalling after spinal cord injury in rat. J Cell Mol Med 2023; 27:482-495. [PMID: 36660907 PMCID: PMC9930430 DOI: 10.1111/jcmm.17620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/12/2022] [Accepted: 10/28/2022] [Indexed: 01/21/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a highly destructive disease in human neurological functions. Adipose-derived mesenchymal stem cells (ADMSCs) have tissue regenerations and anti-inflammations, especially with prion protein overexpression (PrPcOE ). Therefore, this study tested whether PrPcOE -ADMSCs therapy offered benefits in improving outcomes via regulating nod-like-receptor-protein-3 (NLRP3) inflammasome/DAMP signalling after acute SCI in rats. Compared with ADMSCs only, the capabilities of PrPcOE -ADMSCs were significantly enhanced in cellular viability, anti-oxidative stress and migration against H2 O2 and lipopolysaccharide damages. Similarly, PrPcOE -ADMSCs significantly inhibited the inflammatory patterns of Raw264.7 cells. The SD rats (n = 32) were categorized into group 1 (Sham-operated-control), group 2 (SCI), group 3 (SCI + ADMSCs) and group 4 (SCI + PrPcOE -ADMSCs). Compared with SCI group 2, both ADMSCs and PrPcOE -ADMSCs significantly improved neurological functions. Additionally, the circulatory inflammatory cytokines levels (TNF-α/IL-6) and inflammatory cells (CD11b/c+/MPO+/Ly6G+) were highest in group 2, lowest in group 1, and significantly higher in group 3 than in group 4. By Day 3 after SCI induction, the protein expressions of inflammasome signalling (HGMB1/TLR4/MyD88/TRIF/c-caspase8/FADD/p-NF-κB/NEK7/NRLP3/ASC/c-caspase1/IL-ß) and by Day 42 the protein expressions of DAMP-inflammatory signalling (HGMB1/TLR-4/MyD88/TRIF/TRAF6/p-NF-κB/TNF-α/IL-1ß) in spinal cord tissues displayed an identical pattern as the inflammatory patterns. In conclusion, PrPcOE -ADMSCs significantly attenuated SCI in rodents that could be through suppressing the inflammatory signalling.
Collapse
Affiliation(s)
- Tsung‐Cheng Yin
- Department of Orthopaedic SurgeryKaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung UniversityKaohsiungTaiwan,Center for General EducationCheng Shiu UniversityKaohsiungTaiwan
| | - Yi‐Chen Li
- Clinical Medicine Research CenterNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan,Center of Cell TherapyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan,Institute of Clinical MedicineCollege of MedicineNational Cheng Kung UniversityTainanTaiwan,Division of Cardiology, Department of Internal MedicineKaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung UniversityKaohsiungTaiwan
| | - Pei‐Hsun Sung
- Division of Cardiology, Department of Internal MedicineKaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung UniversityKaohsiungTaiwan,Center for Shockwave Medicine and Tissue EngineeringKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan,Institute for Translational Research in BiomedicineKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
| | - John Y. Chiang
- Department of Computer Science & EngineeringNational Sun Yat‐sen UniversityKaohsiungTaiwan,Department of Healthcare Administration and Medical InformaticsKaohsiung Medical UniversityKaohsiungTaiwan
| | - Pei‐Lin Shao
- Department of NursingAsia UniversityTaichungTaiwan
| | - Hon‐Kan Yip
- Division of Cardiology, Department of Internal MedicineKaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung UniversityKaohsiungTaiwan,Center for Shockwave Medicine and Tissue EngineeringKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan,Institute for Translational Research in BiomedicineKaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan,Department of NursingAsia UniversityTaichungTaiwan,Department of Medical ResearchChina Medical University Hospital, China Medical UniversityTaichungTaiwan,Division of Cardiology, Department of Internal MedicineXiamen Chang Gung HospitalXiamenChina
| | - Mel S. Lee
- Department of Orthopaedic SurgeryKaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung UniversityKaohsiungTaiwan,Department of Orthopedic SurgeryPao‐Chien HospitalPingtungTaiwan
| |
Collapse
|
26
|
Amini M, Yousefi Z, Ghafori SS, Hassanzadeh G. Sleep deprivation and NLRP3 inflammasome: Is there a causal relationship? Front Neurosci 2022; 16:1018628. [PMID: 36620464 PMCID: PMC9815451 DOI: 10.3389/fnins.2022.1018628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
In the modern era, sleep deprivation (SD) is one of the most common health problems that has a profound influence on an individual's quality of life and overall health. Studies have identified the possibility that lack of sleep can stimulate inflammatory responses. NLRP3 inflammasome, a key component of the innate immune responses, initiates inflammatory responses by enhancing proinflammatory cytokine release and caspase-1-mediated pyroptosis. In this study, NLRP3 modification, its proinflammatory role, and potential targeted therapies were reviewed with regard to SD-induced outcomes. A growing body of evidence has showed the importance of the mechanistic connections between NLRP3 and the detrimental consequences of SD, but there is a need for more clinically relevant data. In animal research, (i) some animals show differential vulnerability to the effects of SD compared to humans. (ii) Additionally, the effects of sleep differ depending on the SD technique employed and the length of SD. Moreover, paying attention to the crosstalk of all the driving factors of NLRP3 inflammasome activation such as inflammatory responses, autonomic control, oxidative stress, and endothelial function is highly recommended. In conclusion, targeting NLRP3 inflammasome or its downstream pathways for therapy could be complicated due to the reciprocal and complex relationship of SD with NLRP3 inflammasome activation. However, additional research is required to support such a causal claim.
Collapse
Affiliation(s)
- Mohammad Amini
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Sayed Soran Ghafori
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran,*Correspondence: Gholamreza Hassanzadeh,
| |
Collapse
|
27
|
Irandoost E, Najibi S, Talebbeigi S, Nassiri S. Focus on the role of NLRP3 inflammasome in the pathology of endometriosis: a review on molecular mechanisms and possible medical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 396:621-631. [PMID: 36542122 DOI: 10.1007/s00210-022-02365-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Endometriosis (EMS) is a gynecological disease that leads to pathological conditions, which are connected to the initiation of pro-inflammatory cytokine production. Inflammation plays a vital role in the pathogenesis of EMS. The activation and formation of cytoplasmic inflammasome complexes is considered an important step of inflammation and a key regulator of pyroptosis, a form of cell death. NLR family pyrin domain containing 3 (NLRP3) inflammasome complex modulates innate immune activity and inflammation. The NLRP3 inflammasome activates cysteine protease caspase-1, which produces active pro-inflammatory interleukins (ILs), including IL-1β and IL-18. The aim of this review article was to discuss the involvement of NLRP3 inflammasome assembly and its activation in the pathophysiology of EMS and target related pathways in designing appropriate therapeutic approaches. Dysregulation of sex hormone signaling pathways was associated with over-activation of the NLPR3 inflammasome. In this study, we demonstrated the involvement of NLRP3 inflammasome signaling pathways in the pathophysiology of EMS. The manuscript also discusses the beneficial effects of targeted therapy through synthetic inhibitors of NLRP3 signaling pathways to control EMS lesions.
Collapse
Affiliation(s)
- Elnaz Irandoost
- Department of Gynecology and Obstetrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shaparak Najibi
- Department of Gynecology and Obstetrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Talebbeigi
- Department of Gynecology and Obstetrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saina Nassiri
- Department of Gynecology and Obstetrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Yin J, Yin Z, Lai P, Liu X, Ma J. Pyroptosis in Periprosthetic Osteolysis. Biomolecules 2022; 12:biom12121733. [PMID: 36551161 PMCID: PMC9775904 DOI: 10.3390/biom12121733] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Periprosthetic osteolysis (PPO) along with aseptic loosening (AL) caused by wear particles after artificial joint replacement is the key factor in surgical failure and subsequent revision surgery, however, the precise molecular mechanism underlying PPO remains unclear. Aseptic inflammation triggered by metal particles, resulting in the imbalance between bone formation by osteoblasts and bone resorption by osteoclasts may be the decisive factor. Pyroptosis is a new pro-inflammatory pattern of regulated cell death (RCD), mainly mediated by gasdermins (GSDMs) family, among which GSDMD is the best characterized. Recent evidence indicates that activation of NLRP3 inflammasomes and pyroptosis play a pivotal role in the pathological process of PPO. Here, we review the pathological process of PPO, the molecular mechanism of pyroptosis and the interventions to inhibit the inflammation and pyroptosis of different cells during the PPO. Conclusively, this review provides theoretical support for the search for new strategies and new targets for the treatment of PPO by inhibiting pyroptosis and inflammation.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, Shanghai 201600, China
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
| | - Zhaoyang Yin
- Department of Orthopedics, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang 222000, China
| | - Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
- Correspondence: (J.M.); (X.L.)
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, Shanghai 201600, China
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, China
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Correspondence: (J.M.); (X.L.)
| |
Collapse
|
29
|
Formation and activity of NLRP3 inflammasome and histopathological changes in the lung of corpses with COVID-19. J Mol Histol 2022; 53:883-890. [PMID: 36100803 PMCID: PMC9470508 DOI: 10.1007/s10735-022-10101-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 09/02/2022] [Indexed: 12/03/2022]
Abstract
COVID-19 is a contagious disease that attacks many organs but the lungs are the main organs affected. The inflammasome activation results in the exacerbation of inflammatory response in infectious disease. The aim of this study is to investigate the formation and activity of the NLRP3 inflammasome complex and the histopathological changes caused by the coronavirus in the lung of deceased persons with COVID-19. In total, 10 corpses; 5 corpses with no history of any infectious diseases and COVID-19 and 5 corpses with the cause of death of COVID-19 were included in this study. Lung tissue samples were harvested during autopsy under safe conditions. Fresh tissues in each group were used to measure the genes expression and proteins level of NLRP3, ASC, Caspase-1, IL-1β, IL-6 and TNF-α and a routine hematoxylin and eosin staining was performed for histological assessment. Data are represented as the means ± SD. Statistical significance difference was accepted at a p-value less than 5%. The NLRP3, ASC, Caspase-1, IL-1β, IL-6 and TNF-α genes expression and proteins level were elevated in the lung of the COVID-19 group in comparison with the control group. Histological findings presented the increasing number of polymorphonuclear leukocytes, macrophages and also pulmonary fibrosis in the lungs of corpses with the cause of death of COVID-19. High expression of NLRP3 inflammasome components and its relation with the pathophysiology of the coronavirus-infected lung suggested that targeting the NLRP3 inflammasome could be helpful in achieving a more effective treatment in patients with COVID-19.
Collapse
|
30
|
Mokhtari T. Targeting autophagy and neuroinflammation pathways with plant-derived natural compounds as potential antidepressant agents. Phytother Res 2022; 36:3470-3489. [PMID: 35794794 DOI: 10.1002/ptr.7551] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 11/06/2022]
Abstract
Major depressive disorder (MDD) is a life-threatening disease that presents several characteristics. The pathogenesis of depression still remains poorly understood. Moreover, the mechanistic interactions of natural components in treating depression to target autophagy and neuroinflammation are yet to be evaluated. This study overviewed the effects of plant-derived natural components in regulating critical pathways, particularly neuroinflammation and autophagy, associated with depression. A list of natural components, including luteolin, apigenin, hyperforin, resveratrol, salvianolic acid b, isoliquiritin, nobiletin, andrographolide, and oridonin, have been investigated. All peer-reviewed journal articles were searched by Scopus, MEDLINE, PubMed, Web of Science, and Google Scholar using the appropriated keywords, including depression, neuroinflammation, autophagy, plant, natural components, etc. The neuroinflammation and autophagy dysfunction are critically associated with the pathophysiology of depression. Natural components with higher efficiency and lower complications can be used for targeting neuroinflammation and autophagy. These components with different doses showed the beneficial antidepressant properties in rodents. These can modulate autophagy markers, mainly AMPK, LC3II/LC3I ratio, Beclin-1. Moreover, they can regulate the NLRP3 inflammasome, resulting in the suppression of proinflammatory cytokines (e.g., IL-1β and IL-18). Future in vitro and in vivo studies are required to develop novel therapeutic approaches based on plant-derived active components to treat MDD.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
31
|
Involvement of NF-κB/NLRP3 axis in the progression of aseptic loosening of total joint arthroplasties: a review of molecular mechanisms. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:757-767. [PMID: 35377011 DOI: 10.1007/s00210-022-02232-4] [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: 12/20/2021] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Particulate wear debris can trigger pro-inflammatory bone resorption and result in aseptic loosening. This complication remains major postoperative discomforts and complications for patients who underwent total joint arthroplasty. Recent studies have indicated that wear debris-induced aseptic loosening is associated with the overproduction of pro-inflammatory cytokines. The activation of osteoclasts as a result of inflammatory responses is associated with osteolysis. Moreover, stimulation of inflammatory signaling pathways such as the NF-κB/NLRP3 axis results in the production of pro-inflammatory cytokines. In this review, we first summarized the potential inflammatory mechanisms of wear particle-induced peri-implant osteolysis. Then, the therapeutic approaches, e.g., biological inhibitors, herbal products, and stem cells or their derivatives, with the ability to suppress the inflammatory responses, mainly NF-κB/NLRP3 signaling pathways, were discussed. Based on the results, activation of macrophages following inflammatory stimuli, overproduction of pro-inflammatory cytokines, and subsequent differentiation of osteoclasts in the presence of wear particles lead to bone resorption. The activation of NF-κB/NLRP3 signaling pathways within the macrophages stimulates the production of pro-inflammatory cytokines, e.g., IL-1β, IL-6, and TNF-α. According to in vitro and in vivo studies, novel therapeutics significantly promoted osteogenesis, suppressed osteoclastogenesis, and diminished particle-mediated bone resorption. Conclusively, these findings offer that suppressing pro-inflammatory cytokines by regulating both NF-κB and NLRP3 inflammasome represents a novel approach to attenuate wear-particle-related osteolytic diseases.
Collapse
|
32
|
All-Trans Retinoic Acid-Preconditioned Mesenchymal Stem Cells Improve Motor Function and Alleviate Tissue Damage After Spinal Cord Injury by Inhibition of HMGB1/NF-κB/NLRP3 Pathway Through Autophagy Activation. J Mol Neurosci 2022; 72:947-962. [PMID: 35147911 DOI: 10.1007/s12031-022-01977-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/20/2022] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a significant public health issue that imposes numerous burdens on patients and society. Uncontrolled excessive inflammation in the second pathological phase of SCI can aggravate the injury. In this paper, we hypothesized that suppressing inflammatory pathways via autophagy could aid functional recovery, and prevent spinal cord tissue degeneration following SCI. To this end, we examined the effects of intrathecal injection of all-trans retinoic acid (ATRA)-preconditioned bone marrow mesenchymal stem cells (BM-MSCs) (ATRA-MSCs) on autophagy activity and the HMGB1/NF-κB/NLRP3 inflammatory pathway in an SCI rat model. This study demonstrated that SCI increased the expression of Beclin-1 (an autophagy-related gene) and NLRP3 inflammasome components such as NLRP3, ASC, Caspase-1, and pro-inflammatory cytokines IL-1β, IL-18, IL-6, and TNF-α. Additionally, following SCI, the protein levels of key autophagy factors (Beclin-1 and LC3-II) and HMGB1/NF-κB/NLRP3 pathway factors (HMGB1, p-NF-κB, NLRP3, IL-1β, and TNF-α) increased. Our findings indicated that ATRA-MSCs enhanced Beclin-1 and LC3-II levels, regulated the HMGB1/NF-κB/NLRP3 pathway, and inhibited pro-inflammatory cytokines. These factors improved hind limb motor activity and aided in the survival of neurons. Furthermore, ATRA-MSCs demonstrated greater beneficial effects than MSCs in treating spinal cord injury. Overall, ATRA-MSC treatment revealed beneficial effects on the damaged spinal cord by suppressing excessive inflammation and activating autophagy. Further research and investigation of the pathways involved in SCI and the use of amplified stem cells may be beneficial for future clinical use.
Collapse
|
33
|
WU B, LIANG X, ZHAO F, FAN W, LI C, ZHAO B, REN J. Clinic serum levels of Plin5 is therapeutic target of spinal cord injury and Plin5 reduced inflammation in spinal cord injury via silent information regulator 1 dependent inhibition of NLRP3 inflammasome. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.51121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | | | - Wei FAN
- Shanxi Medical University, China
| | | | - Bin ZHAO
- Shanxi Medical University, China
| | - Jie REN
- Shanxi Medical University, China
| |
Collapse
|
34
|
Wang Y, Xu H, Wang J, Yi H, Song Y. Extracellular Vesicles in the Pathogenesis, Treatment, and Diagnosis of Spinal Cord Injury: A Mini-Review. Curr Stem Cell Res Ther 2022; 17:317-327. [PMID: 35352667 DOI: 10.2174/1574888x17666220330005937] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Benefiting from in-depth research into stem cells, extracellular vesicles (EVs), which are byproducts of cells and membrane-wrapped microvesicles (30-120 nm) containing lipids, proteins, and nucleic acids, may cast light on the research and development of therapeutics capable of improving the neurological recovery of spinal cord injury (SCI) animals. However, the mechanistic modes of action for EVs in alleviating the lesion size of SCI remain to be solved, thus presenting a tremendous gap existing in translation from the laboratory to the clinic. OBJECTIVE The purpose of this minireview was to cover a wide range of basic views on EVs involved in SCI treatment, including the effects of EVs on the pathogenesis, treatment, and diagnosis of spinal cord injury. METHODS We searched databases (i.e., PubMed, Web of Science, Scopus, Medline, and EMBASE) and acquired all accessible articles published in the English language within five years. Studies reporting laboratory applications of EVs in the treatment of SCI were included and screened to include studies presenting relevant molecular mechanisms. RESULTS This review first summarized the basic role of EVs in cell communication, cell death, inflammatory cascades, scar formation, neuronal regrowth, and angiogenesis after SCI, thereby providing insights into neuroprotection and consolidated theories for future clinical application of EVs. CONCLUSION EVs participate in an extremely wide range of cell activities, play a critical role in cell communication centring neurons, and are considered potential therapies and biomarkers for SCI. miRNAs are the most abundant nucleic acids shipped by EVs and effluent cytokines, and they may represent important messengers of EVs and important factors in SCI treatment.
Collapse
Affiliation(s)
- Yang Wang
- Department of Orthopaedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University; No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Hualiang Xu
- Department of Orthopaedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University; No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Jian Wang
- Department of Orthopaedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University; No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Hanxiao Yi
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107, YanJiang Road, Haizhu District, Guangzhou, China
| | - Yancheng Song
- Department of Orthopaedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University; No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong Province, China
| |
Collapse
|
35
|
Advanced approaches to regenerate spinal cord injury: The development of cell and tissue engineering therapy and combinational treatments. Biomed Pharmacother 2021; 146:112529. [PMID: 34906773 DOI: 10.1016/j.biopha.2021.112529] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) devastate event that is commonly caused by traumatic or non-traumatic events. The reinnervation of spinal cord axons is hampered through a myriad of devices counting on the damaged myelin, inflammation, glial scar, and defective inhibitory molecules. Unfortunately, an effective treatment to completely repair SCI and improve functional recovery has not been found. In this regard, strategies such as using cells, biomaterials, biomolecules, and drugs have been reported to be effective for SCI recovery. Furthermore, recent advances in combinatorial treatments, which address various aspects of SCI pathophysiology, provide optimistic outcomes for spinal cord regeneration. According to the global importance of SCI, the goal of this article review is to provide an overview of the pathophysiology of SCI, with an emphasis on the latest modes of intervention and current advanced approaches for the treatment of SCI, in conjunction with an assessment of combinatorial approaches in preclinical and clinical trials. So, this article can give scientists and clinicians' clues to help them better understand how to construct preclinical and clinical studies that could lead to a breakthrough in spinal cord regeneration.
Collapse
|
36
|
Stem Cell Secretome for Spinal Cord Repair: Is It More than Just a Random Baseline Set of Factors? Cells 2021; 10:cells10113214. [PMID: 34831436 PMCID: PMC8625005 DOI: 10.3390/cells10113214] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/02/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Hundreds of thousands of people suffer spinal cord injuries each year. The experimental application of stem cells following spinal cord injury has opened a new era to promote neuroprotection and neuroregeneration of damaged tissue. Currently, there is great interest in the intravenous administration of the secretome produced by mesenchymal stem cells in acute or subacute spinal cord injuries. However, it is important to highlight that undifferentiated neural stem cells and induced pluripotent stem cells are able to adapt to the damaged environment and produce the so-called lesion-induced secretome. This review article focuses on current research related to the secretome and the lesion-induced secretome and their roles in modulating spinal cord injury symptoms and functional recovery, emphasizing different compositions of the lesion-induced secretome in various models of spinal cord injury.
Collapse
|
37
|
Extracellular vesicles as novel approaches for the treatment of osteoarthritis: a narrative review on potential mechanisms. J Mol Histol 2021; 52:879-891. [PMID: 34510315 DOI: 10.1007/s10735-021-10017-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is a progressive degeneration of articular cartilage with involvement of synovial membrane, and subchondral bone. Current treatment approaches have focused on controlling the OA symptoms, pain, and inflammation. Recently, cell-based therapies, including the application of stem cells such as mesenchymal stem cells (MSCs), have been introduced for restoration of the articular cartilage. Despite promising outcomes, there are some limitations in the application of MSCs for OA treatment. It has been demonstrated that the regenerative potential of stem cells is related to the production of paracrine factors. Extracellular vehicles (EVs), the main component of cell secretome, are membrane-bounded structures that deliver biologically active agents. The delivery of molecules (e.g., nucleic acids, proteins, and lipids) leads to cell-to-cell communication and the alteration of cell functions. In this review, general characteristics of EVs, as well as their potential mechanisms in the prevention and treatment of OA were considered. Based on in vitro and in vivo studies, EVs have shown to contribute to cartilage regeneration via suppression of degenerative factors and regulation of chondrocyte function in the synthesis of extracellular matrix components. Also, they inhibit the progression of OA or protect the cartilage from degradation via their impact on inflammatory cytokines. The different signaling pathways of EVs against the pathologic features of OA were summarized in this review. According to the results obtained from several investigations, more investigations should be design to prove the safety and effectiveness of EVs in the treatment and prevention of OA progression.
Collapse
|
38
|
Guo S, Redenski I, Levenberg S. Spinal Cord Repair: From Cells and Tissue Engineering to Extracellular Vesicles. Cells 2021; 10:cells10081872. [PMID: 34440641 PMCID: PMC8394921 DOI: 10.3390/cells10081872] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition, often leading to severe motor, sensory, or autonomic nervous dysfunction. As the holy grail of regenerative medicine, promoting spinal cord tissue regeneration and functional recovery are the fundamental goals. Yet, effective regeneration of injured spinal cord tissues and promotion of functional recovery remain unmet clinical challenges, largely due to the complex pathophysiology of the condition. The transplantation of various cells, either alone or in combination with three-dimensional matrices, has been intensively investigated in preclinical SCI models and clinical trials, holding translational promise. More recently, a new paradigm shift has emerged from cell therapy towards extracellular vesicles as an exciting "cell-free" therapeutic modality. The current review recapitulates recent advances, challenges, and future perspectives of cell-based spinal cord tissue engineering and regeneration strategies.
Collapse
Affiliation(s)
- Shaowei Guo
- The First Affiliated Hospital, Shantou University Medical College, Shantou 515041, China
- Correspondence: (S.G.); (S.L.)
| | - Idan Redenski
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa 32000, Israel;
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa 32000, Israel;
- Correspondence: (S.G.); (S.L.)
| |
Collapse
|
39
|
Dutta D, Khan N, Wu J, Jay SM. Extracellular Vesicles as an Emerging Frontier in Spinal Cord Injury Pathobiology and Therapy. Trends Neurosci 2021; 44:492-506. [PMID: 33581883 PMCID: PMC8159852 DOI: 10.1016/j.tins.2021.01.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/28/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are membrane-delimited particles that are secreted by nearly all cell types. EVs mediate crucial physiological functions and pathophysiological processes in the CNS. As carriers of diverse bioactive cargoes (e.g., proteins, lipids, and nucleic acids) that can be modified in response to external stimuli, EVs have emerged as pathological mediators following neurotrauma such as spinal cord injury (SCI). We discuss the roles of endogenous EVs in the CNS as well as crosstalk with peripheral EVs in relation to neurotrauma, with a particular focus on SCI. We then summarize the status of EV-based therapeutic advances in preclinical animal models for these conditions. Finally, we discuss new bioengineering strategies that are poised to enhance CNS-specific therapeutic capabilities of EVs.
Collapse
Affiliation(s)
- Dipankar Dutta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Niaz Khan
- Department of Anesthesiology, and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Junfang Wu
- Department of Anesthesiology, and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
40
|
Khalatbary AR. Stem cell-derived exosomes as a cell free therapy against spinal cord injury. Tissue Cell 2021; 71:101559. [PMID: 34052745 DOI: 10.1016/j.tice.2021.101559] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/01/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022]
Abstract
Recent evidence suggests that stem cell therapy has beneficial effects on spinal cord injury. It was subsequently established that these beneficial effects may be mediated through release of paracrine factors, a kind of extracellular vesicle known as exosomes. Stem cell-secreted nano-sized exosomes have shown great potential to reduce apoptosis and inflammation, enhance angiogenesis, and improve functional behavioral recovery following spinal cord injury. This review summarizes current knowledge about the influence of exosomes derived from stem cells on spinal cord protection and regeneration with their molecular mechanisms after injury.
Collapse
Affiliation(s)
- Ali Reza Khalatbary
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
41
|
Willis CM, Nicaise AM, Hamel R, Pappa V, Peruzzotti-Jametti L, Pluchino S. Harnessing the Neural Stem Cell Secretome for Regenerative Neuroimmunology. Front Cell Neurosci 2020; 14:590960. [PMID: 33250716 PMCID: PMC7674923 DOI: 10.3389/fncel.2020.590960] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence foresees the secretome of neural stem cells (NSCs) to confer superimposable beneficial properties as exogenous NSC transplants in experimental treatments of traumas and diseases of the central nervous system (CNS). Naturally produced secretome biologics include membrane-free signaling molecules and extracellular membrane vesicles (EVs) capable of regulating broad functional responses. The development of high-throughput screening pipelines for the identification and validation of NSC secretome targets is still in early development. Encouraging results from pre-clinical animal models of disease have highlighted secretome-based (acellular) therapeutics as providing significant improvements in biochemical and behavioral measurements. Most of these responses are being hypothesized to be the result of modulating and promoting the restoration of key inflammatory and regenerative programs in the CNS. Here, we will review the most recent findings regarding the identification of NSC-secreted factors capable of modulating the immune response to promote the regeneration of the CNS in animal models of CNS trauma and inflammatory disease and discuss the increased interest to refine the pro-regenerative features of the NSC secretome into a clinically available therapy in the emerging field of Regenerative Neuroimmunology.
Collapse
Affiliation(s)
- Cory M. Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
42
|
Noori L, Arabzadeh S, Mohamadi Y, Mojaverrostami S, Mokhtari T, Akbari M, Hassanzadeh G. Intrathecal administration of the extracellular vesicles derived from human Wharton's jelly stem cells inhibit inflammation and attenuate the activity of inflammasome complexes after spinal cord injury in rats. Neurosci Res 2020; 170:87-98. [PMID: 32717259 DOI: 10.1016/j.neures.2020.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Activation of inflammasome complexes during spinal cord injury (SCI) lead to conversion of pro-inflammatory cytokines, interleukin-1beta (IL-1β) and interleukin-18 (IL-18) to their active form to initiates the neuroinflammation. Mesenchymal stem cells (MSCs) showed anti-inflammatory properties through their extracellular vehicles (EVs). We investigated immunomodulatory potential of human Wharton's jelly mesenchymal stem cells derived extracellular vesicles (hWJ-MSC-EVs) on inflammasome activity one week after SCI in rats. The gene expression and protein level of IL-1β, IL-18, tumor necrosis factor alpha (TNF-α) and caspase1, were assessed by QPCR and western blotting. Immunohistochemistry (IHC) was done to measure the glial fibrillary acidic protein (GFAP) and Nestin expression. Cell death, histological evaluation and hind limb locomotion was studied by TUNEL assay, Nissl staining and Basso, Beattie, Bresnaham (BBB), respectively. Our finding represented that intrathecally administrated of hWJ-MSC-EVs significantly attenuated expression of the examined factors in both mRNA (P < 0.05 and P ≤ 0.01) and protein levels (P < 0.05 and P ≤ 0.01), decreased GFAP and increased Nestin expression (P < 0.05), reduced cell death and revealed the higher number of typical neurons in ventral horn of spinal cord. Consequently, progress in locomotion. We came to the conclusion that hWJ-MSC-EVs has the potential to control the inflammasome activity after SCI in rats. Moreover, EVs stimulated the neural progenitor cells and modulate the astrocyte activity.
Collapse
Affiliation(s)
- Leila Noori
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Arabzadeh
- Department of Biology, School of Basic Sciences, Ale Taha Institute of Higher Education, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Mohammad Akbari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience and addiction studies, School of advanced technologies in medicine, Tehran University of Medical Sciences, Tehran, Iran; Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran.
| |
Collapse
|