1
|
Liu Z, Pan C, Huang H. The role of axon guidance molecules in the pathogenesis of epilepsy. Neural Regen Res 2025; 20:1244-1257. [PMID: 39075893 DOI: 10.4103/nrr.nrr-d-23-01620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/21/2024] [Indexed: 07/31/2024] Open
Abstract
Current treatments for epilepsy can only manage the symptoms of the condition but cannot alter the initial onset or halt the progression of the disease. Consequently, it is crucial to identify drugs that can target novel cellular and molecular mechanisms and mechanisms of action. Increasing evidence suggests that axon guidance molecules play a role in the structural and functional modifications of neural networks and that the dysregulation of these molecules is associated with epilepsy susceptibility. In this review, we discuss the essential role of axon guidance molecules in neuronal activity in patients with epilepsy as well as the impact of these molecules on synaptic plasticity and brain tissue remodeling. Furthermore, we examine the relationship between axon guidance molecules and neuroinflammation, as well as the structural changes in specific brain regions that contribute to the development of epilepsy. Ample evidence indicates that axon guidance molecules, including semaphorins and ephrins, play a fundamental role in guiding axon growth and the establishment of synaptic connections. Deviations in their expression or function can disrupt neuronal connections, ultimately leading to epileptic seizures. The remodeling of neural networks is a significant characteristic of epilepsy, with axon guidance molecules playing a role in the dynamic reorganization of neural circuits. This, in turn, affects synapse formation and elimination. Dysregulation of these molecules can upset the delicate balance between excitation and inhibition within a neural network, thereby increasing the risk of overexcitation and the development of epilepsy. Inflammatory signals can regulate the expression and function of axon guidance molecules, thus influencing axonal growth, axon orientation, and synaptic plasticity. The dysregulation of neuroinflammation can intensify neuronal dysfunction and contribute to the occurrence of epilepsy. This review delves into the mechanisms associated with the pathogenicity of axon guidance molecules in epilepsy, offering a valuable reference for the exploration of therapeutic targets and presenting a fresh perspective on treatment strategies for this condition.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | | | | |
Collapse
|
2
|
Ahmed W, Mushtaq A, Ali S, Khan N, Liang Y, Duan L. Engineering Approaches for Exosome Cargo Loading and Targeted Delivery: Biological versus Chemical Perspectives. ACS Biomater Sci Eng 2024; 10:5960-5976. [PMID: 38940421 DOI: 10.1021/acsbiomaterials.4c00856] [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: 06/29/2024]
Abstract
Exosomes are nanoscale membrane bound vesicles secreted by almost all types of cells. Their unique attributes, such as minimal immunogenicity and compatibility with biological systems, make them novel carriers for drug delivery. These native exosomes harbor proteins, nucleic acids, small molecule compounds, and fluorogenic agents. Moreover, through a combination of chemical and bioengineering methodologies, exosomes are tailored to transport precise therapeutic payloads to designated cells or tissues. In this review, we summarize the strategies for exosome modification and drug loading modalities in engineered exosomes. In addition, we provide an overview of the advances in the use of engineered exosomes for targeted drug delivery. Lastly, we discuss the merits and limitations of chemically engineered versus bioengineered exosome-mediated target therapies. These insights offer additional options for refining engineered exosomes in pharmaceutical development and hold promise for expediting the successful translation of engineered exosomes from the bench to the bedside.
Collapse
Affiliation(s)
- Waqas Ahmed
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
- Medical School, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Asim Mushtaq
- Centre for Future Materials, University of Southern Queensland, Springfield, Queensland 4300, Australia
| | - Shahzad Ali
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
- Medical School, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Nawaz Khan
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
- Medical School, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Institute of Mental Health, Shenzhen Mental Health Center, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen 518020, Guangdong, China
| | - Li Duan
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| |
Collapse
|
3
|
Dong J, Gong Z, Bi H, Yang J, Wang B, Du K, Zhang C, Chen L. BMSC-derived exosomal miR-219-5p alleviates ferroptosis in neuronal cells caused by spinal cord injury via the UBE2Z/NRF2 pathway. Neuroscience 2024; 556:73-85. [PMID: 39084457 DOI: 10.1016/j.neuroscience.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE The aim of this study was to investigate the molecular mechanism of exosomal miR-219-5p derived from bone marrow mesenchymal stem cells (BMSCs) in the treatment of spinal cord injury (SCI). METHODS Basso Beattie Bresnahan (BBB) score and tissue staining were used to assess SCI and neuronal survival in rats. The contents of Fe2+, malondialdehyde (MDA), glutathione (GSH), and superoxide dismutase (SOD) were detected by a kit. The expression levels of ubiquitin-conjugating enzyme E2 Z (UBE2Z), nuclear factor erythroid 2-related Factor 2 (NRF2) and ferroptosis-related proteins were detected by Western blotting. In addition, the ability of BMSC-derived exosomes to inhibit ferroptosis in neuronal cells in rats with SCI was validated by in vivo injection of ferroptosis inhibitors/inducers. RESULTS In this study, we found that miR-219-5p-rich BMSC-derived exosomes inhibited ferroptosis in SCI rats and that the alleviating effect of BMSC-Exos on SCI was achieved by inhibiting the ferroptosis signaling pathway and that NRF2 played a key role in this process. Our study confirmed that BMSC exosome-specific delivery of miR-219-5p can target UBE2Z to regulate its stability and that overexpression of UBE2Z reverses miR-219-5p regulation of NRF2. In addition, in vivo experiments showed that BMSC exosomes alleviated ferroptosis in neuronal SCI progression, and inhibiting the expression of miR-219-5p in BMSCs reduced the alleviating effect of exosomes on ferroptosis in neuronal cells and SCI. CONCLUSION miR-219-5p in BMSC-derived exosomes can repair the injured spinal cord. In addition, miR-219-5p alleviates ferroptosis in neuronal cells induced by SCI through the UBE2Z/NRF2 pathway.
Collapse
Affiliation(s)
- Junjie Dong
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Zhiqiang Gong
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Hangchuan Bi
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jin Yang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Bing Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Kaili Du
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Chunqiang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Lingqiang Chen
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China.
| |
Collapse
|
4
|
Qin X, Zhang X, He X, Xu H, Yao Q, Li Z, Feng Y, Zhong Y, Li Z, Lv G, Wang Y. Neuron-derived Netrin-1 deficiency aggravates spinal cord injury through activating the NF-κB signaling pathway. Heliyon 2024; 10:e37388. [PMID: 39290272 PMCID: PMC11407054 DOI: 10.1016/j.heliyon.2024.e37388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/24/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
Netrin-1 (NTN1) is involved in psychological alterations caused by central nerve system diseases. The primary objective of this research was to investigate whether a deficiency of neuron-derived NTN1 in the remote brain regions affects SCI outcomes. To examine the roles and mechanisms of neuron-derived NTN1 during SCI, Western blots, Nissl staining, immunochemical technique, RNA-sequence, and related behavioral tests were conducted in the study. Our study revealed that mice lacking NTN1 exhibited normal morphological structure of the spinal cords, hippocampus, and neurological function. While neuron-derived NTN1deletion mechanistically disrupted neuronal regeneration and aggregates neuronal apoptosis and ferroptosis in the intermediate phase following SCI. Additionally, neuroinflammation was significantly enhanced in the early phase, which could be related to activation of the NF-κB signaling pathway. Overall, our findings indicate that the deletion of neuron-derived NTN1 leads to the activation of the NF-κB pathway, contributing to the promotion of neuronal apoptosis and ferroptosis, and the pathological progression of SCI.
Collapse
Affiliation(s)
- Xiaojian Qin
- Department of Orthopaedics. The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Xiaolan Zhang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Xiaodong He
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Hui Xu
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Qiannan Yao
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Zifeng Li
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Yayun Feng
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Yichen Zhong
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China
| | - Ziyang Li
- Department of Orthopaedics. The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Gang Lv
- Department of Orthopaedics. The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Yanfeng Wang
- Department of Orthopaedics. The First Affiliated Hospital, China Medical University, Shenyang, 110001, Liaoning Province, China
| |
Collapse
|
5
|
Zhang CH, Lu DC, Liu Y, Wang L, Sethi G, Ma Z. The role of extracellular vesicles in pyroptosis-mediated infectious and non-infectious diseases. Int Immunopharmacol 2024; 138:112633. [PMID: 38986299 DOI: 10.1016/j.intimp.2024.112633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Pyroptosis, a lytic and pro-inflammatory cell death, is important in various pathophysiological processes. Host- and bacteria-derived extracellular vesicles (EVs), as natural nanocarriers messengers, are versatile mediators of intercellular communication between different types of cells. Recently, emerging research has suggested that EVs exhibit multifaceted roles in disease progression by manipulating pyroptosis. This review focuses on new findings concerning how EVs shape disease progression in infectious and non-infectious diseases by regulating pyroptosis. Understanding the characteristics and activity of EVs-mediated pyroptotic death may conducive to the discovery of novel mechanisms and more efficient therapeutic targets in infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Cai-Hua Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Department of Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Chongqing 404100, China
| | - Ding-Ci Lu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ying Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China.
| |
Collapse
|
6
|
Zhang J, Hu D, Li L, Qu D, Shi W, Xie L, Jiang Q, Li H, Yu T, Qi C, Fu H. M2 Microglia-derived Exosomes Promote Spinal Cord Injury Recovery in Mice by Alleviating A1 Astrocyte Activation. Mol Neurobiol 2024; 61:7009-7025. [PMID: 38367135 DOI: 10.1007/s12035-024-04026-6] [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: 10/23/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024]
Abstract
M2 microglia transplantation has previously demonstrated beneficial effects on spinal cord injury (SCI) by regulating neuroinflammation and enhancing neuronal survival. Exosomes (EXOs), secreted by almost all cell types, embody partial functions and properties of their parent cells. However, the effect of M2 microglia-derived EXOs (M2-EXOs) on SCI recovery and the underlying molecular mechanisms remain unclear. In this study, we isolated M2-EXOs and intravenously introduced them into mice with SCI. Considering the reciprocal communication between microglia and astroglia in both healthy and injured central nervous systems (CNSs), we subsequently focused on the influence of M2-EXOs on astrocyte phenotype regulation. Our findings indicated that M2-EXOs promoted neuron survival and axon preservation, reduced the lesion area, inhibited A1 astrocyte activation, and improved motor function recovery in SCI mice. Moreover, they inhibited the nuclear translocation of p65 and the activation of the NF-κB signalling pathway in A1 astrocytes. Therefore, our research suggests that M2-EXOs mitigate the activation of neurotoxic A1 astrocytes by inhibiting the NF-κB signalling pathway, thereby improving spinal tissue preservation and motor function recovery following SCI. This positions M2-EXOs as a promising therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Die Hu
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Liping Li
- Department of Bone Surgery, Qingdao Central Hospital, Qingdao, 266000, China
| | - Di Qu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Weipeng Shi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Lei Xie
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
- Department of Orthopedic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Qi Jiang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Haifeng Li
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Tengbo Yu
- Department of Orthopedic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
- Institute of Sports Medicine and Health, Qingdao University, Qingdao, 266000, China
| | - Chao Qi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
7
|
Ye Z, Zheng Y, Li N, Zhang H, Li Q, Wang X. Repair of spinal cord injury by bone marrow mesenchymal stem cell-derived exosomes: a systematic review and meta-analysis based on rat models. Front Mol Neurosci 2024; 17:1448777. [PMID: 39169950 PMCID: PMC11335736 DOI: 10.3389/fnmol.2024.1448777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Objective This study aims to systematically evaluate the efficacy of bone marrow mesenchymal stem cell-derived exosomes (BMSCs-Exo) in improving spinal cord injury (SCI) to mitigate the risk of translational discrepancies from animal experiments to clinical applications. Methods We conducted a comprehensive literature search up to March 2024 using PubMed, Embase, Web of Science, and Scopus databases. Two researchers independently screened the literature, extracted data, and assessed the quality of the studies. Data analysis was performed using STATA16 software. Results A total of 30 studies were included. The results indicated that BMSCs-Exo significantly improved the BBB score in SCI rats (WMD = 3.47, 95% CI [3.31, 3.63]), inhibited the expression of the pro-inflammatory cytokine TNF-α (SMD = -3.12, 95% CI [-3.57, -2.67]), and promoted the expression of anti-inflammatory cytokines IL-10 (SMD = 2.76, 95% CI [1.88, 3.63]) and TGF-β (SMD = 3.89, 95% CI [3.02, 4.76]). Additionally, BMSCs-Exo significantly reduced apoptosis levels (SMD = -4.52, 95% CI [-5.14, -3.89]), promoted the expression of axonal regeneration markers NeuN cells/field (SMD = 3.54, 95% CI [2.65, 4.42]), NF200 (SMD = 4.88, 95% CI [3.70, 6.05]), and the number of Nissl bodies (SMD = 1.89, 95% CI [1.13, 2.65]), and decreased the expression of astrogliosis marker GFAP (SMD = -5.15, 95% CI [-6.47, -3.82]). The heterogeneity among studies was primarily due to variations in BMSCs-Exo transplantation doses, with efficacy increasing with higher doses. Conclusion BMSCs-Exo significantly improved motor function in SCI rats by modulating inflammatory responses, reducing apoptosis, inhibiting astrogliosis, and promoting axonal regeneration. However, the presence of selection, performance, and detection biases in current animal experiments may undermine the quality of evidence in this study.
Collapse
Affiliation(s)
- Zhongduo Ye
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yukun Zheng
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ningning Li
- Lanzhou Maternal and Child Health Hospital, Lanzhou, China
| | - Huaibin Zhang
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qiangqiang Li
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiong Wang
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
Li Z, Zhou T, Bao Z, Wu M, Mao Y. The Porous SilMA Hydrogel Scaffolds Carrying Dual-Sensitive Paclitaxel Nanoparticles Promote Neuronal Differentiation for Spinal Cord Injury Repair. Tissue Eng Regen Med 2024; 21:809-827. [PMID: 39004636 PMCID: PMC11286913 DOI: 10.1007/s13770-024-00659-9] [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: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND In the intricate pathological milieu post-spinal cord injury (SCI), neural stem cells (NSCs) frequently differentiate into astrocytes rather than neurons, significantly limiting nerve repair. Hence, the utilization of biocompatible hydrogel scaffolds in conjunction with exogenous factors to foster the differentiation of NSCs into neurons has the potential for SCI repair. METHODS In this study, we engineered a 3D-printed porous SilMA hydrogel scaffold (SM) supplemented with pH-/temperature-responsive paclitaxel nanoparticles (PTX-NPs). We analyzed the biocompatibility of a specific concentration of PTX-NPs and its effect on NSC differentiation. We also established an SCI model to explore the ability of composite scaffolds for in vivo nerve repair. RESULTS The physical adsorption of an optimal PTX-NPs dosage can simultaneously achieve pH/temperature-responsive release and commendable biocompatibility, primarily reflected in cell viability, morphology, and proliferation. An appropriate PTX-NPs concentration can steer NSC differentiation towards neurons over astrocytes, a phenomenon that is also efficacious in simulated injury settings. Immunoblotting analysis confirmed that PTX-NPs-induced NSC differentiation occurred via the MAPK/ERK signaling cascade. The repair of hemisected SCI in rats demonstrated that the composite scaffold augmented neuronal regeneration at the injury site, curtailed astrocyte and fibrotic scar production, and enhanced motor function recovery in rat hind limbs. CONCLUSION The scaffold's porous architecture serves as a cellular and drug carrier, providing a favorable microenvironment for nerve regeneration. These findings corroborate that this strategy amplifies neuronal expression within the injury milieu, significantly aiding in SCI repair.
Collapse
Affiliation(s)
- Zhixiang Li
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Tao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Zhengqi Bao
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Min Wu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China.
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
- Anhui Engineering Research Center of Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
9
|
Liu R, Jiang L, Chen Y, Shao J, Chen K, Li X, Lv J, Cai W, Cai H, Zhu Z, Wang C, Zhou K, Huang J, Xiao J, Ni W, Wu C. Ginsenoside-Rh2 Promotes Functional Recovery after Spinal Cord Injury by Enhancing TFEB-Mediated Autophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14727-14746. [PMID: 38907713 DOI: 10.1021/acs.jafc.4c02379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Background: Following spinal cord injury (SCI), autophagy plays a positive role in neuronal protection, whereas pyroptosis triggers an inflammatory response. Ginsenoside-Rh2 (GRh2), known for its neuroprotective effects, is considered a promising drug. However, the exact molecular mechanisms underlying these protective effects remain unclear. Aim of the Study: Explore the therapeutic value of GRh2 in SCI and its potential mechanisms of action. Materials and Methods: An SCI mouse model was established, followed by random grouping and drug treatments under different conditions. Subsequently, the functional recovery of SCI mice after GRh2 treatment was assessed using hematoxylin and eosin, Masson's trichrome, and Nissl staining, footprint analysis, Basso Mouse Scale scoring, and inclined plane tests. The expression levels of relevant indicators in the mice were detected using Western blotting, immunofluorescence, and a quantitative polymerase chain reaction. Network pharmacology analysis was used to identify the relevant signaling pathways through which GRh2 exerts its therapeutic effects. Results: GRh2 promoted functional recovery after SCI. GRh2 significantly inhibits pyroptosis by enhancing autophagy in SCI mice. Simultaneously, the neuroprotective effect of GRh2, achieved through the inhibition of pyroptosis, is partially reversed by 3-methyladenine, an autophagy inhibitor. Additionally, the increase in autophagy induced by GRh2 is mediated by the promotion of transcription factor EB (TFEB) nuclear translocation and dephosphorylation. Partial attenuation of the protective effects of GRh2 was observed after TFEB knockdown. Additionally, GRh2 can modulate the activity of TFEB in mice post-SCI through the EGFR-MAPK signaling pathway, and NSC228155 (an EGFR activator) can partially reverse the effect of GRh2 on the EGFR-MAPK signaling pathway. Conclusions: GRh2 improves functional recovery after SCI by upregulating TFEB-mediated autophagic flux and inhibiting pyroptosis, indicating its potential clinical applicability.
Collapse
Affiliation(s)
- Rongjie Liu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Liting Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Yituo Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Jiaqin Shao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Kongbin Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiang Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Junlei Lv
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Wanta Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxu Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhefan Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenggui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Jinfeng Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenfei Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
| | - Chenyu Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
10
|
Hushmandi K, Saadat SH, Raei M, Aref AR, Reiter RJ, Nabavi N, Taheriazam A, Hashemi M. The science of exosomes: Understanding their formation, capture, and role in cellular communication. Pathol Res Pract 2024; 259:155388. [PMID: 38850846 DOI: 10.1016/j.prp.2024.155388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/06/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
Extracellular vesicles (EVs) serve as a crucial method for transferring information among cells, which is vital in multicellular organisms. Among these vesicles, exosomes are notable for their small size, ranging from 20 to 150 nm, and their role in cell-to-cell communication. They carry lipids, proteins, and nucleic acids between cells. The creation of exosomes begins with the inward budding of the cell membrane, which then encapsulates various macromolecules as cargo. Once filled, exosomes are released into the extracellular space and taken up by target cells via endocytosis and similar processes. The composition of exosomal cargo varies, encompassing diverse macromolecules with specific functions. Because of their significant roles, exosomes have been isolated from various cell types, including cancer cells, endothelial cells, macrophages, and mesenchymal cells, with the aim of harnessing them for therapeutic applications. Exosomes influence cellular metabolism, and regulate lipid, glucose, and glutamine pathways. Their role in pathogenesis is determined by their cargo, which can manipulate processes such as apoptosis, proliferation, inflammation, migration, and other molecular pathways in recipient cells. Non-coding RNA transcripts, a common type of cargo, play a pivotal role in regulating disease progression. Exosomes are implicated in numerous biological and pathological processes, including inflammation, cancer, cardiovascular diseases, diabetes, wound healing, and ischemic-reperfusion injury. As a result, they hold significant potential in the treatment of both cancerous and non-cancerous conditions.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Epidemiology and Biostatistics, School of Health, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Reza Aref
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Tang J, Wang X, Lin X, Wu C. Mesenchymal stem cell-derived extracellular vesicles: a regulator and carrier for targeting bone-related diseases. Cell Death Discov 2024; 10:212. [PMID: 38697996 PMCID: PMC11066013 DOI: 10.1038/s41420-024-01973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
The escalating threat of bone-related diseases poses a significant challenge to human health. Mesenchymal stem cell (MSC)-derived extracellular vesicles (MSC-EVs), as inherent cell-secreted natural products, have emerged as promising treatments for bone-related diseases. Leveraging outstanding features such as high biocompatibility, low immunogenicity, superior biological barrier penetration, and extended circulating half-life, MSC-EVs serve as potent carriers for microRNAs (miRNAs), long no-code RNAs (lncRNAs), and other biomolecules. These cargo molecules play pivotal roles in orchestrating bone metabolism and vascularity through diverse mechanisms, thereby contributing to the amelioration of bone diseases. Additionally, engineering modifications enhance the bone-targeting ability of MSC-EVs, mitigating systemic side effects and bolstering their clinical translational potential. This review comprehensively explores the mechanisms through which MSC-EVs regulate bone-related disease progression. It delves into the therapeutic potential of MSC-EVs as adept drug carriers, augmented by engineered modification strategies tailored for osteoarthritis (OA), rheumatoid arthritis (RA), osteoporosis, and osteosarcoma. In conclusion, the exceptional promise exhibited by MSC-EVs positions them as an excellent solution with considerable translational applications in clinical orthopedics.
Collapse
Affiliation(s)
- Jiandong Tang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xiangyu Wang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xu Lin
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Chao Wu
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China.
| |
Collapse
|
12
|
Ma T, Xu G, Gao T, Zhao G, Huang G, Shi J, Chen J, Song J, Xia J, Ma X. Engineered Exosomes with ATF5-Modified mRNA Loaded in Injectable Thermogels Alleviate Osteoarthritis by Targeting the Mitochondrial Unfolded Protein Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21383-21399. [PMID: 38626424 DOI: 10.1021/acsami.3c17209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1β-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.
Collapse
Affiliation(s)
- Tiancong Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guangyu Xu
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Tian Gao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guanglei Zhao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Gangyong Huang
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jingsheng Shi
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jie Chen
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jian Song
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jun Xia
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Xiaosheng Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| |
Collapse
|
13
|
Cheng LF, You CQ, Peng C, Ren JJ, Guo K, Liu TL. Mesenchymal stem cell-derived exosomes as a new drug carrier for the treatment of spinal cord injury: A review. Chin J Traumatol 2024; 27:134-146. [PMID: 38570272 PMCID: PMC11138942 DOI: 10.1016/j.cjtee.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating traumatic disease seriously impairing the quality of life in patients. Expectations to allow the hopeless central nervous system to repair itself after injury are unfeasible. Developing new approaches to regenerate the central nervous system is still the priority. Exosomes derived from mesenchymal stem cells (MSC-Exo) have been proven to robustly quench the inflammatory response or oxidative stress and curb neuronal apoptosis and autophagy following SCI, which are the key processes to rescue damaged spinal cord neurons and restore their functions. Nonetheless, MSC-Exo in SCI received scant attention. In this review, we reviewed our previous work and other studies to summarize the roles of MSC-Exo in SCI and its underlying mechanisms. Furthermore, we also focus on the application of exosomes as drug carrier in SCI. In particular, it combs the advantages of exosomes as a drug carrier for SCI, imaging advantages, drug types, loading methods, etc., which provides the latest progress for exosomes in the treatment of SCI, especially drug carrier.
Collapse
Affiliation(s)
- Lin-Fei Cheng
- Medical College, Anhui University of Science and Technology, Huainan, 232000, Anhui province, China
| | - Chao-Qun You
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Cheng Peng
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Jia-Ji Ren
- Department of Orthopaedic Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Kai Guo
- Department of Orthopaedics, The Central Hospital of Shanghai Putuo District, Shanghai, 200333, China
| | - Tie-Long Liu
- Medical College, Anhui University of Science and Technology, Huainan, 232000, Anhui province, China.
| |
Collapse
|
14
|
曹 春, 邵 高, 张 铭, 李 波, 徐 海, 张 红, 屈 一. [Therapeutic mechanism of basic fibroblast growth factor on spinal cord injury in rats based on the Notch/signal transducer and activator of transcription 3 signaling pathway]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2024; 38:480-486. [PMID: 38632070 PMCID: PMC11024520 DOI: 10.7507/1002-1892.202312066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024]
Abstract
Objective To explore the therapeutic effect of basic fibroblast growth factor (bFGF) on spinal cord injury (SCI) in rats and the influence of Notch/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Methods A total of 40 10-week-old male Sprague Dawley (SD) rats were selected to establish T 10-segment SCI model by a free falling object. Among them, 32 successful models were randomly divided into model group and bFGF group, with 16 in each group. Another 16 SD rats were selected as sham-operation group, with only T 10 processes, dura mater, and spinal cord exposed. After modeling, the rats in bFGF group were intraperitoneally injected with 100 μg/kg bFGF (once a day for 28 days), and the rats in model group and sham-operation group were injected with normal saline in the same way. The survival of rats in each group were observed after modeling. Basso-Beattie-Bresnahan (BBB) scores were performed before modeling and at immediate, 14 days, and 28 days after modeling to evaluate the functional recovery of hind limbs. Then, the spinal cord tissue at the site of injury was taken at 28 days and stained with HE, Nissl, and propidium iodide (PI) to observe the pathological changes, neuronal survival (number of Nissl bodies) and apoptosis (number of PI red stained cells) of the spinal cord tissue; immunohistochemical staining and ELISA were used to detect the levels of astrocyte activation markers [glial fibrillary acidic protein (GFAP)] and inflammatory factors [interleukin 1β (IL-1β), tumor necrosis factor α (TNF-α), interferon γ (IFN-γ)] in tissues, respectively. Western blot was used to detect the expressions of Notch/STAT3 signaling pathway related proteins [Notch, STAT3, phosphoryl-STAT3 (p-STAT3), bone morphogenetic protein 2 (BMP-2)] in tissues. Results All rats survived until the experiment was completed. At immediate after modeling, the BBB scores in model group and bFGF group significantly decreased when compared to sham-operation group ( P<0.05). At 14 and 28 days after modeling, the BBB scores in model group significantly decreased when compared to sham-operation group ( P<0.05); the bFGF group showed an increase compared to model group ( P<0.05). Compared with before modeling, the BBB scores of model group and bFGF group decreased at immediate after modeling, and gradually increased at 14 and 28 days, the differences between different time points were significant ( P<0.05). The structure of spinal cord tissue in sham-operation group was normal; in model group, there were more necrotic lesions in the spinal cord tissue and fewer Nissl bodies with normal structures; the number of necrotic lesions in the spinal cord tissue of the bFGF group significantly reduced compared to the model group, and some normally structured Nissl bodies were visible. Compared with sham-operation group, the number of Nissl bodies in spinal cord tissue significantly decreased, the number of PI red stained cells, GFAP, IL-1β, TNF-α, IFN-γ, Notch, p-STAT3 /STAT3, BMP-2 protein expression levels significantly increased in model group ( P<0.05). The above indexes in bFGF group significantly improved when compared with model group ( P<0.05). Conclusion bFGF can improve motor function and pathological injury repair of spinal cord tissue in SCI rats, improve neuronal survival, and inhibit neuronal apoptosis, excessive activation of astrocytes in spinal cord tissue and inflammatory response, the mechanism of which may be related to the decreased activity of Notch/STAT3 signaling pathway.
Collapse
Affiliation(s)
- 春风 曹
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| | - 高海 邵
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| | - 铭华 张
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| | - 波 李
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| | - 海涛 徐
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| | - 红军 张
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| | - 一鸣 屈
- 重庆医科大学附属永川医院骨科(重庆 402160)Department of Orthopedics, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, 402160, P. R. China
| |
Collapse
|
15
|
Liu J, Cao J, Yu X, Chang J, Sui T, Cao X. Necroptosis pathway emerged as potential diagnosis markers in spinal cord injury. J Cell Mol Med 2024; 28:e18219. [PMID: 38509743 PMCID: PMC10955161 DOI: 10.1111/jcmm.18219] [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: 10/13/2023] [Revised: 01/15/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
The present research focused on identifying necroptosis-related differentially expressed genes (NRDEGs) in spinal cord injury (SCI) to highlight potential therapeutic and prognostic target genes in clinical SCI. Three SCI-related datasets were downloaded, including GSE151371, GSE5296 and GSE47681. MSigDB and KEGG datasets were searched for necroptosis-related genes (NRGs). Differentially expressed genes (DEGs) and NRGs were intersected to obtain NRDEGs. The MCC algorithm was employed to select the first 10 genes as hub genes. A protein-protein interaction (PPI) network related to NRDEGs was developed utilizing STRING. Several databases were searched to predict interactions between hub genes and miRNAs, transcription factors, potential drugs, and small molecules. Immunoassays were performed to identify DEGs using CIBERSORTx. Additionally, qRT-PCR was carried out to verify NRDEGs in an animal model of SCI. Combined analysis of all datasets identified 15 co-expressed DEGs and NRGs. GO and KEGG pathway analyses highlighted DEGs mostly belonged to pathways associated with necroptosis and apoptosis. Hub gene expression analysis showed high accuracy in SCI diagnosis was associated with the expression of CHMP7 and FADD. A total of two hub genes, i.e. CHMP7, FADD, were considered potential targets for SCI therapy.
Collapse
Affiliation(s)
- Jingcheng Liu
- Department of OrthopedicsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsuChina
| | - Jiang Cao
- Department of OrthopedicsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsuChina
| | - Xiao Yu
- Department of OrthopedicsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsuChina
| | - Jie Chang
- Department of OrthopedicsThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Tao Sui
- Department of OrthopedicsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsuChina
| | - Xiaojian Cao
- Department of OrthopedicsThe First Affiliated Hospital with Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
16
|
Lou J, Jin M, Zhou C, Fan Y, Ni L, Mao Y, Shen H, Li J, Zhang H, Fu C, Mao X, Chen Y, Zhong J, Zhou K, Wang L, Wu J. Ezrin inhibition alleviates oxidative stress and pyroptosis via regulating TRPML1-calcineurin axis mediated enhancement of autophagy in spinal cord injury. Free Radic Biol Med 2024; 212:133-148. [PMID: 38142951 DOI: 10.1016/j.freeradbiomed.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Spinal cord injury (SCI) presents profound ramifications for patients, leading to diminished motor and sensory capabilities distal to the lesion site. Once SCI occurs, it not only causes great physical and psychological problems for patients but also imposes a heavy economic burden. Ezrin is involved in various cellular processes, including signal transduction, cell death, inflammation, chemotherapy resistance and the stress response. However, whether Ezrin regulates functional repair after SCI and its underlying mechanism has not been elucidated. Here, our results showed that there is a marked augmentation of Ezrin levels within neurons and Ezrin inhibition markedly diminished glial scarring and bolstered functional recuperation after SCI. RNA sequencing indicated the potential involvement of pyroptosis, oxidative stress and autophagy in the enhancement of functional recovery upon reduced Ezrin expression. Moreover, the inhibition of Ezrin expression curtailed pyroptosis and oxidative stress by amplifying autophagy. Our studies further demonstrated that Ezrin inhibition promoted autophagy by increasing TFEB activity via the Akt-TRPML1-calcineurin pathway. Finally, we concluded that inhibiting Ezrin expression alleviates pyroptosis and oxidative stress by enhancing TFEB-driven autophagy, thereby promoting functional recovery after SCI, which may be a promising therapeutic target for SCI treatment.
Collapse
Affiliation(s)
- Junsheng Lou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Mengran Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Conghui Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Yunpeng Fan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Libin Ni
- Department of Orthopaedics, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, No. 1229 Gudun Road, Hangzhou, 310030, Zhejiang, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Honghao Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Jiafeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Haojie Zhang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Chunyan Fu
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xingjia Mao
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yingying Chen
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Linlin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Tarim University, School of Medicine, Alaer, 843300, China.
| | - Junsong Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
17
|
Morishima Y, Kawabori M, Yamazaki K, Takamiya S, Yamaguchi S, Nakahara Y, Senjo H, Hashimoto D, Masuda S, Fujioka Y, Ohba Y, Mizuno Y, Kuge Y, Fujimura M. Intravenous Administration of Mesenchymal Stem Cell-Derived Exosome Alleviates Spinal Cord Injury by Regulating Neutrophil Extracellular Trap Formation through Exosomal miR-125a-3p. Int J Mol Sci 2024; 25:2406. [PMID: 38397083 PMCID: PMC10889446 DOI: 10.3390/ijms25042406] [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: 01/21/2024] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Spinal cord injury (SCI) leads to devastating sequelae, demanding effective treatments. Recent advancements have unveiled the role of neutrophil extracellular traps (NETs) produced by infiltrated neutrophils in exacerbating secondary inflammation after SCI, making it a potential target for treatment intervention. Previous research has established that intravenous administration of stem cell-derived exosomes can mitigate injuries. While stem cell-derived exosomes have demonstrated the ability to modulate microglial reactions and enhance blood-brain barrier integrity, their impact on neutrophil deactivation, especially in the context of NETs, remains poorly understood. This study aims to investigate the effects of intravenous administration of MSC-derived exosomes, with a specific focus on NET formation, and to elucidate the associated molecular mechanisms. Exosomes were isolated from the cell supernatants of amnion-derived mesenchymal stem cells using the ultracentrifugation method. Spinal cord injuries were induced in Sprague-Dawley rats (9 weeks old) using a clip injury model, and 100 μg of exosomes in 1 mL of PBS or PBS alone were intravenously administered 24 h post-injury. Motor function was assessed serially for up to 28 days following the injury. On Day 3 and Day 28, spinal cord specimens were analyzed to evaluate the extent of injury and the formation of NETs. Flow cytometry was employed to examine the formation of circulating neutrophil NETs. Exogenous miRNA was electroporated into neutrophil to evaluate the effect of inflammatory NET formation. Finally, the biodistribution of exosomes was assessed using 64Cu-labeled exosomes in animal positron emission tomography (PET). Rats treated with exosomes exhibited a substantial improvement in motor function recovery and a reduction in injury size. Notably, there was a significant decrease in neutrophil infiltration and NET formation within the spinal cord, as well as a reduction in neutrophils forming NETs in the circulation. In vitro investigations indicated that exosomes accumulated in the vicinity of the nuclei of activated neutrophils, and neutrophils electroporated with the miR-125a-3p mimic exhibited a significantly diminished NET formation, while miR-125a-3p inhibitor reversed the effect. PET studies revealed that, although the majority of the transplanted exosomes were sequestered in the liver and spleen, a notably high quantity of exosomes was detected in the damaged spinal cord when compared to normal rats. MSC-derived exosomes play a pivotal role in alleviating spinal cord injury, in part through the deactivation of NET formation via miR-125a-3p.
Collapse
Affiliation(s)
- Yutaka Morishima
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan; (Y.M.); (K.Y.); (S.T.); (M.F.)
| | - Masahito Kawabori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan; (Y.M.); (K.Y.); (S.T.); (M.F.)
| | - Kazuyoshi Yamazaki
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan; (Y.M.); (K.Y.); (S.T.); (M.F.)
| | - Soichiro Takamiya
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan; (Y.M.); (K.Y.); (S.T.); (M.F.)
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe 650-0047, Hyogo, Japan
| | - Yo Nakahara
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan; (Y.M.); (K.Y.); (S.T.); (M.F.)
| | - Hajime Senjo
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Daigo Hashimoto
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Sakiko Masuda
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Hokkaido, Japan;
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan
| | - Yuki Mizuno
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Hokkaido, Japan; (Y.M.)
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Hokkaido, Japan; (Y.M.)
| | - Miki Fujimura
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Hokkaido, Japan; (Y.M.); (K.Y.); (S.T.); (M.F.)
| |
Collapse
|
18
|
Xu Y, Geng Z, Sun Y, Zhu G, Xiao L, Wang Z, Li B, Liu X, Shi J, Song X, Hu J, Qi Q. Complanatuside A improves functional recovery after spinal cord injury through inhibiting JNK signaling-mediated microglial activation. Eur J Pharmacol 2024; 965:176287. [PMID: 38158110 DOI: 10.1016/j.ejphar.2023.176287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AND AIMS Complanatuside A (ComA) is a flavonoid-rich compound in Astragalus membranaceus that has anti-inflammatory and neuroprotective effects. In this study, we focused on the effect of ComA on spinal cord injury (SCI) in mice and explored its possible mechanisms. METHODS The SCI model was constructed using C57BL/6J mice, and the effect of ComA on motor function recovery in SCI mice was evaluated through the BMS (Basso Mouse Scale) and footprint test. The histological effects of ComA on SCI mice were evaluated by hematoxylin-eosin (H&E) staining, Luxol-fast blue (LFB) staining, and Nissl staining. In both in vivo and in vitro experiments, we detected the activation of microglia and the release of inflammatory factors through molecular experiments. Immunofluorescence and Western blotting confirmed that ComA can prevent neuronal apoptosis caused by activated microglia through the c-Jun N-terminal kinase (JNK) pathway. RESULTS Our research results confirm that ComA can improve motor function in mice after SCI. Our in vitro results indicate that ComA can inhibit the activation of BV2 cells and the release of proinflammatory mediators. In addition, ComA can prevent neuronal cell apoptosis caused by activated BV2 cells. Finally, we found that ComA works through the JNK signaling pathway. CONCLUSIONS ComA can accelerate the restoration of motor function in mice after SCI, possibly by reducing neuronal apoptosis via inhibition of JNK-related signaling pathways, a reduction in microglial activation, and inhibition of inflammatory factor release. Our data indicate that ComA is a promising drug candidate for improving functional recovery in patients with SCI.
Collapse
Affiliation(s)
- Yibo Xu
- School of Basic Medicine, Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yang Sun
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Guoqing Zhu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Linyu Xiao
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Zhiyuan Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Bohan Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Xinyue Liu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Jinran Shi
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| | - Qi Qi
- School of Basic Medicine, Bengbu Medical University, Bengbu, Anhui, China; Key Laboratory of Tissue Transplantation, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
19
|
Jang HJ, Shim KS, Lee J, Park JH, Kang SJ, Shin YM, Lee JB, Baek W, Yoon JK. Engineering of Cell Derived-Nanovesicle as an Alternative to Exosome Therapy. Tissue Eng Regen Med 2024; 21:1-19. [PMID: 38066355 PMCID: PMC10764700 DOI: 10.1007/s13770-023-00610-4] [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: 10/04/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Exosomes, nano-sized vesicles ranging between 30 and 150 nm secreted by human cells, play a pivotal role in long-range intercellular communication and have attracted significant attention in the field of regenerative medicine. Nevertheless, their limited productivity and cost-effectiveness pose challenges for clinical applications. These issues have recently been addressed by cell-derived nanovesicles (CDNs), which are physically synthesized exosome-mimetic nanovesicles from parent cells, as a promising alternative to exosomes. CDNs exhibit structural, physical, and biological properties similar to exosomes, containing intracellular protein and genetic components encapsulated by the cell plasma membrane. These characteristics allow CDNs to be used as regenerative medicine and therapeutics on their own, or as a drug delivery system. METHODS The paper reviews diverse methods for CDN synthesis, current analysis techniques, and presents engineering strategies to improve lesion targeting efficiency and/or therapeutic efficacy. RESULTS CDNs, with their properties similar to those of exosomes, offer a cost-effective and highly productive alternative due to their non-living biomaterial nature, nano-size, and readiness for use, allowing them to overcome several limitations of conventional cell therapy methods. CONCLUSION Ongoing research and enhancement of CDNs engineering, along with comprehensive safety assessments and stability analysis, exhibit vast potential to advance regenerative medicine by enabling the development of efficient therapeutic interventions.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Kyu-Sik Shim
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jinah Lee
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Joo Hyeon Park
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seong-Jun Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Young Min Shin
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jung Bok Lee
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Wooyeol Baek
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea.
| |
Collapse
|
20
|
Tian J, Han Z, Song D, Peng Y, Xiong M, Chen Z, Duan S, Zhang L. Engineered Exosome for Drug Delivery: Recent Development and Clinical Applications. Int J Nanomedicine 2023; 18:7923-7940. [PMID: 38152837 PMCID: PMC10752020 DOI: 10.2147/ijn.s444582] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/16/2023] [Indexed: 12/29/2023] Open
Abstract
Exosomes are nano-sized membrane vesicles that transfer bioactive molecules between cells and modulate various biological processes under physiological and pathological conditions. By applying bioengineering technologies, exosomes can be modified to express specific markers or carry therapeutic cargo and emerge as novel platforms for the treatment of cancer, neurological, cardiovascular, immune, and infectious diseases. However, there are many challenges and uncertainties in the clinical translation of exosomes. This review aims to provide an overview of the recent advances and challenges in the translation of engineered exosomes, with a special focus on the methods and strategies for loading drugs into exosomes, the pros and cons of different loading methods, and the optimization of exosome production based on the drugs to be encapsulated. Moreover, we also summarize the current clinical applications and prospects of engineered exosomes, as well as the potential risks and limitations that need to be addressed in exosome engineering, including the standardization of exosome preparation and engineering protocols, the quality and quantity of exosomes, the control of drug release, and the immunogenicity and cytotoxicity of exosomes. Overall, engineered exosomes represent an exciting frontier in nanomedicine, but they still face challenges in large-scale production, the maintenance of storage stability, and clinical translation. With continuous advances in this field, exosome-based drug formulation could offer great promise for the targeted treatment of human diseases.
Collapse
Affiliation(s)
- Jiaqi Tian
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
| | - Zhengpu Han
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
- School of Public Health, Weifang Medical University, Weifang, People’s Republic of China
| | - Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
| | - Yanjie Peng
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
| | - Min Xiong
- School of Public Health, North China University of Science and Technology, Tangshan, People’s Republic of China
| | - Zhen Chen
- School of Public Health, Weifang Medical University, Weifang, People’s Republic of China
| | - Shuyin Duan
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Lin Zhang
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
- Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Jinan, People’s Republic of China
| |
Collapse
|
21
|
Li Y, Li YJ, Zhu ZQ. To re-examine the intersection of microglial activation and neuroinflammation in neurodegenerative diseases from the perspective of pyroptosis. Front Aging Neurosci 2023; 15:1284214. [PMID: 38020781 PMCID: PMC10665880 DOI: 10.3389/fnagi.2023.1284214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and motor neuron disease, are diseases characterized by neuronal damage and dysfunction. NDs are considered to be a multifactorial disease with diverse etiologies (immune, inflammatory, aging, genetic, etc.) and complex pathophysiological processes. Previous studies have found that neuroinflammation and typical microglial activation are important mechanisms of NDs, leading to neurological dysfunction and disease progression. Pyroptosis is a new mode involved in this process. As a form of programmed cell death, pyroptosis is characterized by the expansion of cells until the cell membrane bursts, resulting in the release of cell contents that activates a strong inflammatory response that promotes NDs by accelerating neuronal dysfunction and abnormal microglial activation. In this case, abnormally activated microglia release various pro-inflammatory factors, leading to the occurrence of neuroinflammation and exacerbating both microglial and neuronal pyroptosis, thus forming a vicious cycle. The recognition of the association between pyroptosis and microglia activation, as well as neuroinflammation, is of significant importance in understanding the pathogenesis of NDs and providing new targets and strategies for their prevention and treatment.
Collapse
Affiliation(s)
- Yuan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- College of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Ying-Jie Li
- Department of General Surgery, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Zhao-Qiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
22
|
Li D, Lu X, Xu G, Liu S, Gong Z, Lu F, Xia X, Jiang J, Wang H, Zou F, Ma X. Dihydroorotate dehydrogenase regulates ferroptosis in neurons after spinal cord injury via the P53-ALOX15 signaling pathway. CNS Neurosci Ther 2023. [PMID: 36942513 DOI: 10.1111/cns.14150] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a highly disabling condition in spinal surgery that leads to neuronal damage and secondary inflammation. Ferroptosis is a non-apoptotic type of cell death that has only recently been identified, which is marked primarily by iron-dependent and lipid-derived reactive oxygen species accumulation, and accompanied by morphological modifications such as mitochondrial atrophy and increase in membrane density. Dihydroorotate dehydrogenase (DHODH) is a powerful inhibitor of ferroptosis and has been demonstrated to inhibit cellular ferroptosis in tumor cells, but whether it can inhibit neuronal injury following spinal cord injury remains ambiguous. METHODS In this study, the effect of DHODH on neuronal ferroptosis was observed in vivo and in vitro using a rat spinal cord injury model and erastin-induced PC12 cells, respectively. A combination of molecular and histological approaches was performed to assess ferroptosis and explore the possible mechanisms in vivo and in vitro. RESULTS First, we confirmed the existence of neuronal ferroptosis after spinal cord injury and that DHODH attenuates neuronal damage after spinal cord injury. Second, we showed molecular evidence that DHODH inhibits the activation of ferroptosis-related molecules and reduces lipid peroxide production and mitochondrial damage, thereby reducing neuronal ferroptosis. Further analysis suggests that P53/ALOX15 may be one of the mechanisms regulated by DHODH. Importantly, we determined that DHODH inhibits ALOX15 expression by inhibiting P53. CONCLUSIONS Our findings reveal a novel function for DHODH in neuronal ferroptosis after spinal cord injury, suggesting a unique therapeutic target to alleviate the disease process of spinal cord injury.
Collapse
Affiliation(s)
- Dachuan Li
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiao Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Guangyu Xu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Siyang Liu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaoyang Gong
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Feizhou Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianyuan Jiang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongli Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Fei Zou
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaosheng Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|