1
|
Sterner RC, Sterner RM. Immune response following traumatic spinal cord injury: Pathophysiology and therapies. Front Immunol 2023; 13:1084101. [PMID: 36685598 PMCID: PMC9853461 DOI: 10.3389/fimmu.2022.1084101] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating condition that is often associated with significant loss of function and/or permanent disability. The pathophysiology of SCI is complex and occurs in two phases. First, the mechanical damage from the trauma causes immediate acute cell dysfunction and cell death. Then, secondary mechanisms of injury further propagate the cell dysfunction and cell death over the course of days, weeks, or even months. Among the secondary injury mechanisms, inflammation has been shown to be a key determinant of the secondary injury severity and significantly worsens cell death and functional outcomes. Thus, in addition to surgical management of SCI, selectively targeting the immune response following SCI could substantially decrease the progression of secondary injury and improve patient outcomes. In order to develop such therapies, a detailed molecular understanding of the timing of the immune response following SCI is necessary. Recently, several studies have mapped the cytokine/chemokine and cell proliferation patterns following SCI. In this review, we examine the immune response underlying the pathophysiology of SCI and assess both current and future therapies including pharmaceutical therapies, stem cell therapy, and the exciting potential of extracellular vesicle therapy.
Collapse
Affiliation(s)
- Robert C. Sterner
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Rosalie M. Sterner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States,*Correspondence: Rosalie M. Sterner,
| |
Collapse
|
2
|
0.1% RGN-259 (Thymosin ß4) Ophthalmic Solution Promotes Healing and Improves Comfort in Neurotrophic Keratopathy Patients in a Randomized, Placebo-Controlled, Double-Masked Phase III Clinical Trial. Int J Mol Sci 2022; 24:ijms24010554. [PMID: 36613994 PMCID: PMC9820614 DOI: 10.3390/ijms24010554] [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: 11/21/2022] [Revised: 12/15/2022] [Accepted: 12/18/2022] [Indexed: 12/30/2022] Open
Abstract
We determined the efficacy and safety of 0.1% RGN-259 ophthalmic solution (containing the regenerative protein thymosin ß4) in promoting the healing of persistent epithelial defects in patients with Stages 2 and 3 neurotrophic keratopathy. Complete healing occurred after 4 weeks in 6 of the 10 RGN-259-treated subjects and in 1 of the 8 placebo-treated subjects (p = 0.0656), indicating a strong efficacy trend. Additional efficacy was seen in the significant healing (p = 0.0359) with no recurrent defects observed at day 43, two weeks after cessation of treatment, while the one healed placebo-treated subject at day 28 suffered a recurrence at day 43. The Mackie classification disease stage improved in the RGN-259-treated group at Days 29, 36, and 43 (p = 0.0818, 0.0625, and 0.0467, respectively). Time to complete healing also showed a trend towards efficacy (p = 0.0829, Kaplan-Meier) with 0.1% RGN-259. RGN-259-treated subjects had significant improvements at multiple time points in ocular discomfort, foreign body sensation, and dryness which were not seen in the placebo group. No significant adverse effects were observed. In summary, the use of 0.1% RGN-259 promotes rapid healing of epithelial defects in neurotrophic keratopathy, improves ocular comfort, and is safe for treating this challenging population of patients.
Collapse
|
3
|
Hellenbrand DJ, Quinn CM, Piper ZJ, Morehouse CN, Fixel JA, Hanna AS. Inflammation after spinal cord injury: a review of the critical timeline of signaling cues and cellular infiltration. J Neuroinflammation 2021; 18:284. [PMID: 34876174 PMCID: PMC8653609 DOI: 10.1186/s12974-021-02337-2] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/30/2021] [Indexed: 03/02/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating neurological condition that results in a loss of motor and sensory function. Although extensive research to develop treatments for SCI has been performed, to date, none of these treatments have produced a meaningful amount of functional recovery after injury. The primary injury is caused by the initial trauma to the spinal cord and results in ischemia, oxidative damage, edema, and glutamate excitotoxicity. This process initiates a secondary injury cascade, which starts just a few hours post-injury and may continue for more than 6 months, leading to additional cell death and spinal cord damage. Inflammation after SCI is complex and driven by a diverse set of cells and signaling molecules. In this review, we utilize an extensive literature survey to develop the timeline of local immune cell and cytokine behavior after SCI in rodent models. We discuss the precise functional roles of several key cytokines and their effects on a variety of cell types involved in the secondary injury cascade. Furthermore, variations in the inflammatory response between rats and mice are highlighted. Since current SCI treatment options do not successfully initiate functional recovery or axonal regeneration, identifying the specific mechanisms attributed to secondary injury is critical. With a more thorough understanding of the complex SCI pathophysiology, effective therapeutic targets with realistic timelines for intervention may be established to successfully attenuate secondary damage.
Collapse
Affiliation(s)
- Daniel J Hellenbrand
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Charles M Quinn
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Zachariah J Piper
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Carolyn N Morehouse
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Jordyn A Fixel
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Amgad S Hanna
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA.
| |
Collapse
|
4
|
Wang M, Feng LR, Li ZL, Ma KG, Chang KW, Chen XL, Yang PB, Ji SF, Ma YB, Han H, Ruganzua JB, Yang WN, Qian YH. Thymosin β4 reverses phenotypic polarization of glial cells and cognitive impairment via negative regulation of NF-κB signaling axis in APP/PS1 mice. J Neuroinflammation 2021; 18:146. [PMID: 34183019 PMCID: PMC8240373 DOI: 10.1186/s12974-021-02166-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/04/2021] [Indexed: 01/15/2023] Open
Abstract
Background Thymosin β4 (Tβ4) is the most abundant member of the β-thymosins and plays an important role in the control of actin polymerization in eukaryotic cells. While its effects in multiple organs and diseases are being widely investigated, the safety profile has been established in animals and humans, currently, little is known about its influence on Alzheimer’s disease (AD) and the possible mechanisms. Thus, we aimed to evaluate the effects and mechanisms of Tβ4 on glial polarization and cognitive performance in APP/PS1 transgenic mice. Methods Behavior tests were conducted to assess the learning and memory, anxiety and depression in APP/PS1 mice. Thioflavin S staining, Nissl staining, immunohistochemistry/immunofluorescence, ELISA, qRT-PCR, and immunoblotting were performed to explore Aβ accumulation, phenotypic polarization of glial cells, neuronal loss and function, and TLR4/NF-κB axis in APP/PS1 mice. Results We demonstrated that Tβ4 protein level elevated in all APP/PS1 mice. Over-expression of Tβ4 alone alleviated AD-like phenotypes of APP/PS1 mice, showed less brain Aβ accumulation and more Insulin-degrading enzyme (IDE), reversed phenotypic polarization of microglia and astrocyte to a healthy state, improved neuronal function and cognitive behavior performance, and accidentally displayed antidepressant-like effect. Besides, Tβ4 could downregulate both TLR4/MyD88/NF-κB p65 and p52-dependent inflammatory pathways in the APP/PS1 mice. While combination drug of TLR4 antagonist TAK242 or NF-κB p65 inhibitor PDTC exerted no further effects. Conclusions These results suggest that Tβ4 may exert its function by regulating both classical and non-canonical NF-κB signaling and is restoring its function as a potential therapeutic target against AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02166-3.
Collapse
Affiliation(s)
- Meng Wang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Li-Rong Feng
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zi-Long Li
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kai-Ge Ma
- Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ke-Wei Chang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xin-Lin Chen
- Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Peng-Bo Yang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Sheng-Feng Ji
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yan-Bing Ma
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hua Han
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - John Bosco Ruganzua
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wei-Na Yang
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yi-Hua Qian
- Department of Human Anatomy and Histology-Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China. .,Institute of Neuroscience, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China. .,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
5
|
Al Mamun A, Monalisa I, Tul Kubra K, Akter A, Akter J, Sarker T, Munir F, Wu Y, Jia C, Afrin Taniya M, Xiao J. Advances in immunotherapy for the treatment of spinal cord injury. Immunobiology 2020; 226:152033. [PMID: 33321368 DOI: 10.1016/j.imbio.2020.152033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/19/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a leading cause of morbidity and disability in the world. Over the past few decades, the exact molecular mechanisms describing secondary, persistent injuries, as well as primary and transient injuries, have attracted massive attention to the clinicians and researchers. Recent investigations have distinctly shown the critical roles of innate and adaptive immune responses in regulating sterile neuroinflammation and functional outcomes after SCI. In past years, some promising advances in immunotherapeutic options have efficaciously been identified for the treatment of SCI. In our narrative review, we have mainly focused on the new therapeutic strategies such as the maturation and apoptosis of immune cells by several agents, mesenchymal stem cells (MSCs) as well as multi-factor combination therapy, which have recently provided novel ideas and prospects for the future treatment of SCI. This article also illustrates the latest progress in clarifying the potential roles of innate and adaptive immune responses in SCI, the progression and specification of prospective immunotherapy and outstanding issues in the area.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China
| | - Ilma Monalisa
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Khadija Tul Kubra
- Department of Pharmacy, University of Development Alternative, Dhaka 1209, Bangladesh
| | - Afroza Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Jaheda Akter
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chattogram-4318, Chittagong, Bangladesh
| | - Tamanna Sarker
- Department of Pharmacy, University of Asia Pacific, Dhaka 1205, Bangladesh
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035 Zhejiang Province, China
| | - Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027 Zhejiang Province, China
| | - Masuma Afrin Taniya
- Department of Life Sciences, School of Environment and Life Sciences, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China.
| |
Collapse
|
6
|
Zhang GH, Murthy KD, Binti Pare R, Qian YH. Protective effect of Tβ4 on central nervous system tissues and its developmental prospects. EUR J INFLAMM 2020. [DOI: 10.1177/2058739220934559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tissue repair and regeneration in the central nervous system (CNS) remains a serious medical problem. CNS diseases such as traumatic and neurological brain injuries have a high mortality and disability rate, thereby bringing a considerable amount of economic burden to society and families. How to treat traumatic and neurological brain injuries has always been a serious issue faced by neurosurgeons. The global incidence of traumatic and neurological brain injuries has gradually increased and become a global challenge. Thymosin β4 (Tβ4) is the main G-actin variant molecule in eukaryotic cells. During the development of the CNS, Tβ4 regulates neurogenesis, tangential expansion, tissue growth, and cerebral hemisphere folding. In addition, Tβ4 has anti-apoptotic and anti-inflammatory properties. It promotes angiogenesis, wound healing, stem/progenitor cell differentiation, and other characteristics of cell migration and survival, providing a scientific basis for the repair and regeneration of injured nerve tissue. This review provides evidence to support the role of Tβ4 in the protection and repair of nervous tissue in CNS diseases, especially with the potential to control brain inflammatory processes, and thus open up new therapeutic applications for a series of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gui-hong Zhang
- School of Medicine, Xi’an International University, Xi’an, China
- Department of Biomedical Science and Therapeutic, Faculty of Medicine and Health Sciences (FPSK), Universiti Malaysia Sabah (UMS), Kota Kinabalu, Malaysia
| | - Krishna Dilip Murthy
- Department of Biomedical Science and Therapeutic, Faculty of Medicine and Health Sciences (FPSK), Universiti Malaysia Sabah (UMS), Kota Kinabalu, Malaysia
| | - Rahmawati Binti Pare
- Department of Biomedical Science and Therapeutic, Faculty of Medicine and Health Sciences (FPSK), Universiti Malaysia Sabah (UMS), Kota Kinabalu, Malaysia
| | - Yi-hua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| |
Collapse
|
7
|
Mohammed I, Ijaz S, Mokhtari T, Gholaminejhad M, Mahdavipour M, Jameie B, Akbari M, Hassanzadeh G. Subventricular zone-derived extracellular vesicles promote functional recovery in rat model of spinal cord injury by inhibition of NLRP3 inflammasome complex formation. Metab Brain Dis 2020; 35:809-818. [PMID: 32185593 DOI: 10.1007/s11011-020-00563-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 03/05/2020] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is the destruction of spinal cord motor and sensory resulted from an attack on the spinal cord, which can cause significant physiological damage. The inflammasome is a multiprotein oligomer resulting in inflammation; the NLRP3 inflammasome composed of NLRP3, apoptosis-associated speck-like protein (ASC), procaspase-1, and cleavage of procaspase-1 into caspase-1 initiates the inflammatory response. Subventricular Zone (SVZ) is the origin of neural stem/progenitor cells (NS/PCs) in the adult brain. Extracellular vesicles (EVs) are tiny lipid membrane bilayer vesicles secreted by different types of cells playing an important role in cell-cell communications. The aim of this study was to investigate the effect of intrathecal transplantation of EVs on the NLRP3 inflammasome formation in SCI rats. Male wistar rats were divided into three groups as following: laminectotomy group, SCI group, and EVs group. EVs was isolated from SVZ, and characterized by western blot and DLS, and then injected into the SCI rats. Real-time PCR and western blot were carried out for gene expression and protein level of NLRP3, ASC, and Caspase-1. H&E and cresyl violet staining were performed for histological analyses, as well as BBB test for motor function. The results indicated high level in mRNA and protein level in SCI group in comparison with laminectomy (p < 0.001), and injection of EVs showed a significant reduction in the mRNA and protein levels in EVs group compared to SCI (p < 0.001). H&E and cresyl violet staining showed recovery in neural cells of spinal cord tissue in EVs group in comparison with SCI group. BBB test showed the promotion of motor function in EVs group compared to SCI in 14 days (p < 0.05). We concluded that the injection of EVs could recover the motor function in rats with SCI and rescue the neural cells of spinal cord tissue by suppressing the formation of the NLRP3 inflammasome complex.
Collapse
Affiliation(s)
- Ibrahim Mohammed
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Ijaz
- 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
| | - Morteza Gholaminejhad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Mahdavipour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnamedin Jameie
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - 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.
| |
Collapse
|
8
|
Much More Than a Scaffold: Cytoskeletal Proteins in Neurological Disorders. Cells 2020; 9:cells9020358. [PMID: 32033020 PMCID: PMC7072452 DOI: 10.3390/cells9020358] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
Recent observations related to the structure of the cytoskeleton in neurons and novel cytoskeletal abnormalities involved in the pathophysiology of some neurological diseases are changing our view on the function of the cytoskeletal proteins in the nervous system. These efforts allow a better understanding of the molecular mechanisms underlying neurological diseases and allow us to see beyond our current knowledge for the development of new treatments. The neuronal cytoskeleton can be described as an organelle formed by the three-dimensional lattice of the three main families of filaments: actin filaments, microtubules, and neurofilaments. This organelle organizes well-defined structures within neurons (cell bodies and axons), which allow their proper development and function through life. Here, we will provide an overview of both the basic and novel concepts related to those cytoskeletal proteins, which are emerging as potential targets in the study of the pathophysiological mechanisms underlying neurological disorders.
Collapse
|
9
|
Chio JCT, Wang J, Badner A, Hong J, Surendran V, Fehlings MG. The effects of human immunoglobulin G on enhancing tissue protection and neurobehavioral recovery after traumatic cervical spinal cord injury are mediated through the neurovascular unit. J Neuroinflammation 2019; 16:141. [PMID: 31288834 PMCID: PMC6615094 DOI: 10.1186/s12974-019-1518-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/11/2019] [Indexed: 12/30/2022] Open
Abstract
Background Spinal cord injury (SCI) is a condition with few effective treatment options. The blood-spinal cord barrier consists of pericytes, astrocytes, and endothelial cells, which are collectively termed the neurovascular unit. These cells support spinal cord homeostasis by expressing tight junction proteins. Physical trauma to the spinal cord disrupts the barrier, which leads to neuroinflammation by facilitating immune cell migration to the damaged site in a process involving immune cell adhesion. Immunosuppressive strategies, including methylprednisolone (MPSS), have been investigated to treat SCI. However, despite some success, MPSS has the potential to increase a patient’s susceptibility to wound infection and impaired wound healing. Hence, immunomodulation may be a more attractive approach than immunosuppression. Approved for modulating neuroinflammation in certain disorders, including Guillain-Barre syndrome, intravenous administration of human immunoglobulin G (hIgG) has shown promise in the setting of experimental SCI, though the optimal dose and mechanism of action remain undetermined. Methods Female adult Wistar rats were subjected to moderate-severe clip compression injury (35 g) at the C7-T1 level and randomized to receive a single intravenous (IV) bolus of hIgG (0.02, 0.2, 0.4, 1, 2 g/kg), MPSS (0.03 g/kg), or control buffer at 15 min post-SCI. At 24 h and 6 weeks post-SCI, molecular, histological, and neurobehavioral effects of hIgG were analyzed. Results At 24 h post-injury, human immunoglobulin G co-localized with spinal cord pericytes, astrocytes, and vessels. hIgG (2 g/kg) protected the spinal cord neurovasculature after SCI by increasing tight junction protein expression and reducing inflammatory enzyme expression. Improvements in vascular integrity were associated with changes in spinal cord inflammation. Interestingly, hIgG (2 g/kg) increased serum expression of inflammatory cytokines and co-localized (without decreasing protein expression) with spinal cord vascular cell adhesion molecule-1, a protein used by immune cells to enter into inflamed tissue. Acute molecular benefits of hIgG (2 g/kg) led to greater tissue preservation, functional blood flow, and neurobehavioral recovery at 6 weeks post-SCI. Importantly, the effects of hIgG (2 g/kg) were superior to control buffer and hIgG (0.4 g/kg), and comparable with MPSS (0.03 g/kg). Conclusions hIgG (2 g/kg) is a promising therapeutic approach to mitigate secondary pathology in SCI through antagonizing immune cell infiltration at the level of the neurovascular unit.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jian Wang
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada
| | - Anna Badner
- Sue and Bill Gross Stem Cell Research Centre, University of California, 845 Health Sciences Road, Irvine, CA, 92617, USA
| | - James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | | | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, 7KD-430, Toronto, Ontario, M5T 2S8, Canada. .,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada. .,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada. .,University of Toronto, Toronto, Ontario, Canada. .,Gerry and Tootsie Halbert Chair in Neural Repair and Regeneration, University of Toronto, Toronto, Canada. .,Krembil Neuroscience Program, Toronto Western Hospital, University Health Network, 399 Bathurst Street, Toronto, Ontario, M5T 2S8, Canada.
| |
Collapse
|
10
|
Li H, Wang Y, Hu X, Ma B, Zhang H. Thymosin beta 4 attenuates oxidative stress-induced injury of spinal cord-derived neural stem/progenitor cells through the TLR4/MyD88 pathway. Gene 2019; 707:136-142. [PMID: 31054361 DOI: 10.1016/j.gene.2019.04.083] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 01/22/2023]
Abstract
Neural stem/progenitor cells (NSPCs) can enhance regeneration after spinal cord injury (SCI), but survival of transplanted cells remains poor. Understanding how NSPCs respond to the chemical mediators of secondary injury thus is essential for treating SCI. Thymosin β4 (Tβ4) has physiological functions that are highly relevant to SCI. We exposed NSPCs to oxidative stress and found reduced expression of Tβ4 in H2O2-injured NSPCs. Using an MTT assay, we found that Tβ4 dose dependently increased viability of the injured NSPCs. Tβ4 also reversed the decreases of intracellular Ca2+ concentration and increases of lactate dehydrogenase in NSPCs induced by H2O2 treatment. H2O2 exposure increased NSPC apoptosis, which Tβ4 decreased. In H2O2-induced NSPCs, ROS production and pro-inflammatory cytokines increased, and again, Tβ4 reversed these effects. We investigated the toll-like receptor 4 (TLR4) and myeloid differentiation primary response 88 (MyD88) signaling pathway as an underlying mechanism in Tβ4's protective effect on H2O2-exposed NSPCs. Our results showed that Tβ4 reduced expression of TLR4 and MyD88. Moreover, H2O2-exposed NSPCs that were treated with the TLR4/MyD88 pathway inhibitor showed a reversal of all the effects caused by H2O2, similar to Tβ4's effects. In conclusion, our study determined that Tβ4 attenuated H2O2-induced oxidative stress injury in NSPCs via the TLR4/MyD88 pathway.
Collapse
Affiliation(s)
- Hongwei Li
- Department of Spine SurgAery, Lanzhou University Second Hospital, Lanzhou 740030, Gansu, China
| | - Yonggang Wang
- Department of Spine SurgAery, Lanzhou University Second Hospital, Lanzhou 740030, Gansu, China
| | - Xuchang Hu
- Department of Spine SurgAery, Lanzhou University Second Hospital, Lanzhou 740030, Gansu, China
| | - Bing Ma
- Department of Spine SurgAery, Lanzhou University Second Hospital, Lanzhou 740030, Gansu, China
| | - Haihong Zhang
- Department of Spine SurgAery, Lanzhou University Second Hospital, Lanzhou 740030, Gansu, China.
| |
Collapse
|
11
|
Osei J, Kelly W, Toffolo K, Donahue K, Levy B, Bard J, Wang J, Levy E, Nowak N, Poulsen D. Thymosin beta 4 induces significant changes in the plasma miRNA profile following severe traumatic brain injury in the rat lateral fluid percussion injury model. Expert Opin Biol Ther 2019; 18:159-164. [PMID: 29873258 DOI: 10.1080/14712598.2018.1484102] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Thymosin beta 4 (Tβ4) has demonstrated neuroprotective potential in models of neurlogical injury. The neuroprotective potential of Tβ4 has been associated with increased miR-200a and miR-200b within the brain following stroke. Here we tested the hypothesis that Tβ4 treatment could also alter miRNA profiles within the plasma following severe traumatic brain injury (TBI). METHODS We used the rat lateral fluid percusion injury model of severe TBI to test this hypothesis. Highly sensitive and quantitative droplet digital polymerase chain reaction (ddPCR) was used to measure the plasma concentrations of miR-200 family members. In addition, we conducted RNAseq analysis of plasma miRNA to further identify changes associated with TBI and treatment with Tβ4. RESULTS ddPCR demonstrated that miR-200a-3p andmiR-200b-3p were both significantly increased in plasma following treatment with Tβ4 after severe TBI. RNAseq analysis suggested that miR-300-3p and miR-598-3p increased while miR-450-3p and miR-194-5p significantly decreased following TBI. In contrast, miR-194-5p significantly increased in Tβ4 treated rats following TBI. In addition, we identified nine plasma miRNAs whose expression significantly changed following treatment with Tβ4. CONCLUSIONS Tβ4 treatment significantly increased plasma levels of miR-200a-3p and miR-200b-3p, while RNAseq analysis identified miR-194-5p as a candidate miRNA that may be critical for neuroprotection.
Collapse
Affiliation(s)
- Jennifer Osei
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| | - William Kelly
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| | - Kathryn Toffolo
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| | - Kaitlynn Donahue
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| | - Bennet Levy
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| | - Jonathan Bard
- b New York State Center for Bioinformatics and Life Sciences , University at Buffalo , Buffalo , NY , USA
| | - Jianxin Wang
- c Center for Computational Research , University at Buffalo , Buffalo , NY , USA
| | - Elad Levy
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| | - Norma Nowak
- b New York State Center for Bioinformatics and Life Sciences , University at Buffalo , Buffalo , NY , USA.,d Department of Biochemistry, School of Medicine and Biomedical Sciences , Univeristy at Buffalo , Buffalo , NY , USA
| | - David Poulsen
- a Department of Neurosrgery, Jacobs School of Medicine and Biomedical Sceinces , Univeristy at Buffalo , Buffalo , NY , USA
| |
Collapse
|
12
|
Sinomenine Attenuates Traumatic Spinal Cord Injury by Suppressing Oxidative Stress and Inflammation via Nrf2 Pathway. Neurochem Res 2019; 44:763-775. [DOI: 10.1007/s11064-018-02706-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/18/2018] [Accepted: 12/22/2018] [Indexed: 12/15/2022]
|
13
|
Severa M, Zhang J, Giacomini E, Rizzo F, Etna MP, Cruciani M, Garaci E, Chopp M, Coccia EM. Thymosins in multiple sclerosis and its experimental models: moving from basic to clinical application. Mult Scler Relat Disord 2019; 27:52-60. [PMID: 30317071 PMCID: PMC7104151 DOI: 10.1016/j.msard.2018.09.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) afflicts more than 2.5 million individuals worldwide and this number is increasing over time. Within the past years, a great number of disease-modifying treatments have emerged; however, efficacious treatments and a cure for MS await discovery. Thymosins, soluble hormone-like peptides produced by the thymus gland, can mediate immune and non-immune physiological processes and have gained interest in recent years as therapeutics in inflammatory and autoimmune diseases. METHODS Pubmed was searched with no time constraints for articles using a combination of the keywords "thymosin/s" or "thymus factor/s" AND "multiple sclerosis", mesh terms with no language restriction. RESULTS Here, we review the state-of-the-art on the effects of thymosins on MS and its experimental models. In particular, we describe what is known in this field on the roles of thymosin-α1 (Tα1) and -β4 (Tβ4) as potential anti-inflammatory as well as neuroprotective and remyelinating molecules and their mechanisms of action. CONCLUSION Based on the data that Tα1 and Tβ4 act as anti-inflammatory molecules and as inducers of myelin repair and neuronal protection, respectively, a possible therapeutic application in MS for Tα1 and Tβ4 alone or combined with other approved drugs may be envisaged. This approach is reasonable in light of the current clinical usage of Tα1 and data demonstrating the safety, tolerability and efficacy of Tβ4 in clinical practice.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Jing Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA,Department of Physics, Oakland University, Rochester, MI, USA
| | | |
Collapse
|
14
|
Celastrol inhibits microglial pyroptosis and attenuates inflammatory reaction in acute spinal cord injury rats. Int Immunopharmacol 2018; 66:215-223. [PMID: 30472522 DOI: 10.1016/j.intimp.2018.11.029] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022]
Abstract
Pyroptosis pathway is closely related to inflammation. However, Celastrol effect on pyroptosis pathway after spinal cord injury (SCI) are poorly understood. We studied the anti-inflammatory and neuroprotective effects of Celastrol on acute spinal cord injury in rats, and its anti-inflammatory effects on lipopolysaccharide (LPS)/ATP-induced microgliosis. Our results show that Celastrol can improve the recovery of hindlimb motor function after SCI in Sprague-Dawley (SD) rats, and reduce the cavity area of spinal cord injury along with the neuronal loss. Celastrol simultaneously reduced the activation of microglia (especially M1 microglia) in the spinal cord, inhibited the pyroptosis-related proteins (NLRP3 ASC Caspase-1 GSDMD), reduced the release of TNF-α IL-1β and IL-18 inflammatory factors, and increased the release of IL10 cytokines. In vitro studies showed that Celastrol reduced the toxicity resulting from the administration of LPS with ATP to BV-2 cells, inhibited the pyroptosis-related proteins (NLRP3 Caspase-1 GSDMD), and inhibited the release of corresponding inflammatory factors. Finally, Celastrol can inhibit the expression of NFκB/p-p65 in vitro and in vivo. Our results show that Celastrol can attenuate the inflammatory response of the spinal cord after SCI, which is associated with inhibition of microglial activation and pyroptosis pathway. Further study to explore the use of Celastrol to treat SCI is warranted.
Collapse
|
15
|
Jo MJ, Kumar H, Joshi HP, Choi H, Ko WK, Kim JM, Hwang SSS, Park SY, Sohn S, Bello AB, Kim KT, Lee SH, Zeng X, Han I. Oral Administration of α-Asarone Promotes Functional Recovery in Rats With Spinal Cord Injury. Front Pharmacol 2018; 9:445. [PMID: 29867457 PMCID: PMC5949368 DOI: 10.3389/fphar.2018.00445] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/17/2018] [Indexed: 11/13/2022] Open
Abstract
α-asarone, a bioactive compound found in Acorus plant species, has been shown to exhibit neuroprotective, anti-oxidative, anti-inflammatory, and cognitive-enhancing effects. However, the effects of α-asarone on spinal cord injury (SCI) have not yet been elucidated. The present study investigated the effects of α-asarone on the mRNA of pro-inflammatory cytokines, macrophage polarization toward an anti-inflammatory M2 phenotype, and angiogenesis in rats with compressive SCI. α-Asarone was orally administered (10 mg/kg) once per day for 14 days following moderate static compression SCI. Compared to controls, α-asarone treatment significantly improved locomotor score, prevented neuroinflammation, and facilitated angiogenesis in the spinal cord at 14 days after SCI. Furthermore, α-asarone significantly reduced the TNF-α, IL-1β, IL-6, monocyte chemoattractant protein 1 (MCP-1), macrophage inflammatory protein 2 (MIP-2), and inducible nitric oxide synthase (iNOS) levels but increased the IL-4, IL-10, and arginase 1 levels at 24 h after SCI. At 7 and 14 days after SCI, immunohistochemistry showed reduced reactive gliosis and neuroinflammation and an increased expression of M2 macrophage markers and angiogenesis. The results suggest that the inhibition of pro-inflammatory cytokines, macrophage polarization toward an anti-inflammatory M2 phenotype, and angiogenesis by α-asarone may be some of the mechanisms underlying the α-asarone-mediated neuroprotective effects on an injured spinal cord.
Collapse
Affiliation(s)
- Min-Jae Jo
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Hemant Kumar
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Hari P. Joshi
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Wan-Kyu Ko
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - J. M. Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Sean S. S. Hwang
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Song Y. Park
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| | - Alvin B. Bello
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Kyoung-Tae Kim
- Department of Neurosurgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, South Korea
| |
Collapse
|
16
|
Jiang W, Li M, He F, Zhou S, Zhu L. Targeting the NLRP3 inflammasome to attenuate spinal cord injury in mice. J Neuroinflammation 2017; 14:207. [PMID: 29070054 PMCID: PMC5657095 DOI: 10.1186/s12974-017-0980-9] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/12/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a devastating disease, which results in tissue loss and neurologic dysfunction. NLRP3 inflammasome plays an important role in the mechanism of diverse diseases. However, no studies have demonstrated the role of NLRP3 inflammasome and the effects of NLRP3 inflammasome inhibitors in a mouse model of SCI. We investigated whether inhibition of NLRP3 inflammasome activation by the pharmacologic inhibitor BAY 11-7082 or A438079 could exert neuroprotective effects in a mouse model of SCI. METHODS SCI was performed using an aneurysm clip with a closing force of 30 g at the level of the T6-T7 vertebra for 1 min. Motor recovery was evaluated by an open-field test. Neuronal death was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining. Mitochondrial dysfunction was determined by quantitative real-time polymerase chain reaction (qPCR), western blot, and detection of mitochondrial membrane potential level. Microglia/macrophage activation and astrocytic response were evaluated by immunofluorescence labeling. RESULTS Inhibition of NLRP3 inflammasome activation by pharmacologic inhibitor BAY 11-7082 or A438079 reduced neuronal death, attenuated spinal cord anatomic damage, and promoted motor recovery. Furthermore, BAY 11-7082 or A438079 directly attenuated the levels of NLRP3 inflammasome and proinflammatory cytokines. Moreover, BAY 11-7082 or A438079 alleviated microglia/macrophage activation, neutrophils infiltration, and reactive gliosis, as well as mitochondrial dysfunction. CONCLUSIONS Collectively, our results demonstrate that pharmacologic suppression of NLRP3 inflammasome activation controls neuroinflammation, attenuates mitochondrial dysfunction, alleviates the severity of spinal cord damage, and improves neurological recovery after SCI. These data strongly indicate that the NLRP3 inflammasome is a vital contributor to the secondary damage of SCI in mice.
Collapse
Affiliation(s)
- Wu Jiang
- Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China
| | - Maoqiang Li
- Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China
| | - Fan He
- Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China
| | - Shaobo Zhou
- Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China
| | - Liulong Zhu
- Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Shangcheng District, Hangzhou, 310006, China.
| |
Collapse
|
17
|
Zheng XY, Lv YF, Li S, Li Q, Zhang QN, Zhang XT, Hao ZM. Recombinant adeno-associated virus carrying thymosin β 4 suppresses experimental colitis in mice. World J Gastroenterol 2017; 23:242-255. [PMID: 28127198 PMCID: PMC5236504 DOI: 10.3748/wjg.v23.i2.242] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 10/04/2016] [Accepted: 11/13/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the protective effect of a recombinant adeno-associated virus carrying thymosin β4 (AAV-Tβ4) on murine colitis via intracolonic administration.
METHODS AAV-Tβ4 was prepared and intracolonically used to mediate the secretory expression of Tβ4 in mouse colons. Dextran sulfate sodium (DSS) was applied to induce the murine ulcerative colitis, and 2,4,6-trinitrobenzene sulfonic acid (TNBS) was used to establish a mouse colitis model resembling Crohn’s disease. The disease severity and colon injuries were observed and graded to reveal the effects of AAV-Tβ4 on colitis. The activities of myeloperoxidase (MPO) and superoxide dismutase (SOD) and the content of malondialdehyde (MDA) were determined using biochemical assays. Colonic levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-10 were measured using ELISA, and mucosal epithelial cell apoptosis and proliferation were detected by TUNEL assay and immunochemistry, respectively.
RESULTS Recombinant AAVs efficiently delivered LacZ and Tβ4 into the colonic tissues of the mice, and AAV-Tβ4 led to a strong expression of Tβ4 in mouse colons. In both the DSS and TNBS colitis models, AAV-Tβ4-treated mice displayed distinctly attenuated colon injuries and reduced apoptosis rate of colonic mucosal epithelia. AAV-Tβ4 significantly reduced inflammatory cell infiltrations and relieved oxidative stress in the inflamed colons of the mice, as evidenced by decreases in MPO activity and MDA content and increases in SOD activity. AAV-Tβ4 also modulated colonic TNF-α, IL-1β and IL-10 levels and suppressed the compensatory proliferation of colonic epithelial cells in DSS- and TNBS-treated mice.
CONCLUSION Tβ4 exerts a protective effect on murine colitis, indicating that AAV-Tβ4 could potentially be developed into a promising agent for the therapy of inflammatory bowel diseases.
Collapse
|
18
|
Thymosin β4 overexpression regulates neuron production and spatial distribution in the developing avian optic tectum. Histochem Cell Biol 2016; 147:555-564. [DOI: 10.1007/s00418-016-1529-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2016] [Indexed: 12/11/2022]
|
19
|
Mingomataj EÇ, Bakiri AH. Regulator Versus Effector Paradigm: Interleukin-10 as Indicator of the Switching Response. Clin Rev Allergy Immunol 2016; 50:97-113. [PMID: 26450621 DOI: 10.1007/s12016-015-8514-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The interleukin-10 (IL-10) is generally considered as the most important cytokine with anti-inflammatory properties and one of the key cytokines preventing inflammation-mediated tissue damage. In this respect, IL-10 producing cells play a crucial role in the outcome of infections, allergy, autoimmune reactions, tumor development, and transplant tolerance. Based on recent findings with regard to the mentioned clinical conditions, this review attempts to shed some light on the IL-10 functions, considering this cytokine as inherent inducer of the switching immunity. While acute infections and vaccinations are associated by IL-10 enhanced during few weeks, chronic parasitoses, tumor diseases, allergen-specific immunotherapy, transplants, and use of immune-suppressor drugs show an increased IL-10 level along months or years. With regard to autoimmune pathologies, the IL-10 increase is prevalently observed during early stages, whereas the successive stages are characterized by reaching of immune equilibrium independently to disease's activity. Together, these findings indicate that IL-10 is mainly produced during transient immune conditions and the persistent IL-10-related effect is the indication/prediction (and maybe effectuation) of the switching immunity. Actual knowledge emphasizes that any manipulation of the IL-10 response for treatment purposes should be considered very cautiously due to its potential hazards to the immune system. Probably, the IL-10 as potential switcher of immunity response should be used in association with co-stimulatory immune effectors that are necessary to determine the appropriate deviation during treatment of respective pathologies. Hopefully, further findings would open new avenues to study the biology of this "master switch" cytokine and its therapeutic potential.
Collapse
Affiliation(s)
- Ervin Ç Mingomataj
- Department of Allergy & Clinical Immunology, "Mother Theresa" School of Medicine, Tirana, Albania. .,Faculty of Technical Medical Sciences, Department of Preclinical Disciplines, University of Medicine, Tirana, Albania.
| | - Alketa H Bakiri
- Hygeia Hospital Tirana, Outpatients Service, Allergology Consulting Room, Tirana, Albania.,Faculty of Medical Sciences, Department of Preclinical Disciplines, Albanian University, Tirana, Albania
| |
Collapse
|
20
|
Cheng P, Kuang F, Ju G. Aescin reduces oxidative stress and provides neuroprotection in experimental traumatic spinal cord injury. Free Radic Biol Med 2016; 99:405-417. [PMID: 27596954 DOI: 10.1016/j.freeradbiomed.2016.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 12/22/2022]
Abstract
Aescin has many physiological functions that are highly relevant to spinal cord injury (SCI), including anti-inflammation, anti-oxidation, anti-oedema, and enhancing vascular tone. The present study investigated the putative therapeutic value of aescin in SCI, with a focus on its neuroprotective, anti-inflammatory, and anti-oxidative properties. Sodium aescinate (1.0mg/kg body weight) or equivalent volume of saline was administered 30min after injury by intravenous injection, with an additional dose daily for seven consecutive days after moderate SCI in rats. After contusion injury of the 8th thoracic (T8) spinal cord, aescin-treated rats developed less severe hind limb weakness than saline controls, as assayed by the Basso-Beattie-Bresnahan scale, the beam walking test, and a footprint analysis. The improved locomotor outcomes in aescin-treated rats corresponded to markedly decreased immune response, oxidative stress, neuronal loss, axon demyelination, spinal cord swelling, and cell apoptosis, measured around T8 after impact. Our data suggest aescin treatment as a novel, early, neuroprotective approach in SCI. Given the known safety of aescin in clinical applications, the results of this study suggest that it is a good candidate for SCI treatment in humans.
Collapse
Affiliation(s)
- Peng Cheng
- Institute of Neurosciences, PLA Fourth Military Medical University, 169 West Changle Road, Xi'an 710032, China; Department of Neurology, PLA 425th Hospital, 86 Sanya Bay Road, Sanya 572000, China.
| | - Fang Kuang
- Institute of Neurosciences, PLA Fourth Military Medical University, 169 West Changle Road, Xi'an 710032, China
| | - Gong Ju
- Institute of Neurosciences, PLA Fourth Military Medical University, 169 West Changle Road, Xi'an 710032, China.
| |
Collapse
|
21
|
Thymosin β4 impeded murine stem cell proliferation with an intact cardiovascular differentiation. ACTA ACUST UNITED AC 2016; 36:328-334. [PMID: 27376799 DOI: 10.1007/s11596-016-1587-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/30/2016] [Indexed: 12/22/2022]
Abstract
Thymosin β4 (Tβ4) is a key factor in cardiac development, growth, disease, epicardial integrity, blood vessel formation and has cardio-protective properties. However, its role in murine embryonic stem cells (mESCs) proliferation and cardiovascular differentiation remains unclear. Thus we aimed to elucidate the influence of Tβ4 on mESCs. Target genes during mESCs proliferation and differentiation were detected by real-time PCR or Western blotting, and patch clamp was applied to characterize the mESCs-derived cardiomyocytes. It was found that Tβ4 decreased mESCs proliferation in a partial dose-dependent manner and the expression of cell cycle regulatory genes c-myc, c-fos and c-jun. However, mESCs self-renewal markers Oct4 and Nanog were elevated, indicating the maintenance of self-renewal ability in these mESCs. Phosphorylation of STAT3 and Akt was inhibited by Tβ4 while the expression of RAS and phosphorylation of ERK were enhanced. No significant difference was found in BMP2/BMP4 or their downstream protein smad. Wnt3 and Wnt11 were remarkably decreased by Tβ4 with upregulation of Tcf3 and constant β-catenin. Under mESCs differentiation, Tβ4 treatment did not change the expression of cardiovascular cell markers α-MHC, PECAM, and α-SMA. Neither the electrophysiological properties of mESCs-derived cardiomyocytes nor the hormonal regulation by Iso/Cch was affected by Tβ4. In conclusion, Tβ4 suppressed mESCs proliferation by affecting the activity of STAT3, Akt, ERK and Wnt pathways. However, Tβ4 did not influence the in vitro cardiovascular differentiation.
Collapse
|
22
|
Abstract
No agent has been identified that significantly accelerates the repair of chronic dermal wounds in humans. Thymosin beta 4 (Tβ4) is a small, abundant, naturally occurring regenerative protein that is found in body fluids and inside cells. It was found to have angiogenic and antiinflammatory activity and to be high in platelets that aggregate at the wound site. Thus we used Tβ4 initially in dermal healing. It has since been shown to have many activities important in tissue protection, repair, and regeneration. Tβ4 increases the rate of dermal healing in various preclinical animal models, including diabetic and aged animals, and is active for burns as well. Tβ4 also accelerated the rate of repair in phase 2 trials with patients having pressure ulcers, stasis ulcers, and epidermolysis bullosa wounds. It is safe and well tolerated and will likely have additional uses in the skin and in injured organs for tissue repair and regeneration.
Collapse
Affiliation(s)
- H K Kleinman
- George Washington University, Washington, DC, United States.
| | - G Sosne
- Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
23
|
Zhang J, Feng G, Bao G, Xu G, Sun Y, Li W, Wang L, Chen J, Jin H, Cui Z. Nuclear translocation of PKM2 modulates astrocyte proliferation via p27 and -catenin pathway after spinal cord injury. Cell Cycle 2015; 14:2609-18. [PMID: 26151495 PMCID: PMC4613169 DOI: 10.1080/15384101.2015.1064203] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/07/2015] [Accepted: 06/15/2015] [Indexed: 12/27/2022] Open
Abstract
Aberrant functionality of the cell cycle has been implicated in the pathology of traumatic SCI. Although it has been reported that the expressions of various cell cycle related proteins were altered significantly following SCI, detailed information on the subject remains largely unclear. The embryonic pyruvate kinase M2 (PKM2) is an important metabolic kinase in aerobic glycolysis or the warburg effect, however, its functions in central nervous system (CNS) injury remains elusive. Here we demonstrate that PKM2 was not only significantly upregulated by western blot and immunohistochemistry but certain traumatic stimuli also induced translocation of PKM2 into the nucleus in astrocytes following spinal cord injury (SCI). Furthermore, the expression levels and localization of p-β-catenin, p27, cyclin D1 and PCNA were correlated with PKM2 after SCI. In vitro, we also found that PKM2 co-immunoprecipitation with p-β-catenin and p27 respectively. Knockdown of PKM2 apparently decreased the level of PCNA, cyclinD1, p27 in primary astrocyte cells. Taken together, our findings indicate that nuclear translocation of PKM2 promotes astrocytes proliferation after SCI through modulating cell cycle signaling. These discoveries firstly uncovered the role of PKM2 in spinal cord injury and provided a potential therapeutic target for CNS injury and repair.
Collapse
Affiliation(s)
- Jinlong Zhang
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Guijuan Feng
- Department of Stomatology; Affiliated Hospital of Nantong University, Nantong; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Guofeng Bao
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Guanhua Xu
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Yuyu Sun
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Weidong Li
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Lingling Wang
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Jiajia Chen
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Huricha Jin
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| | - Zhiming Cui
- Department of Spine Surgery; The Second Affiliated Hospital of Nantong University; Nantong University; 226001, Nantong, Jiangsu, PR, China
| |
Collapse
|