1
|
Tian X, Le N, Zhao Y, Alawamleh D, Schwartz A, Meyer L, Helm E, Wu C. Upregulating ANKHD1 in PS19 mice reduces Tau phosphorylation and mitigates Tau-toxicity-induced cognitive deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623890. [PMID: 39605390 PMCID: PMC11601383 DOI: 10.1101/2024.11.15.623890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Abnormal accumulation of Tau protein in the brain disrupts normal cellular function and leads to neuronal death linked with many neurodegenerative disorders such as Alzheimer's disease. An attractive approach to mitigate Tau-induced neurodegeneration is to enhance the clearance of toxic Tau aggregates. We previously showed that upregulation of the fly gene mask protects against FUS- and Tau-induced photoreceptor degeneration in fly disease models. Here we have generated a transgenic mouse line carrying Cre-inducible ANKHD1, the mouse homolog of mask, to determine whether the protective role of mask is conserved from flies to mammals. Utilizing the TauP301S-PS19 mouse model for Tau-related dementia, we observed that ANKHD1 significantly reduced hyperphosphorylated human Tau in 6-month-old mice. Additionally, there was a notable trend towards reduced gliosis levels in these mice, suggesting a protective role of ANKHD1 against TauP301S-linked degeneration. Further analysis of 9-month-old mice revealed a similar trend of effects. Moreover, we found that ANKHD1 also suppresses the cognitive defect of 9-month-old PS19 female mice in novel object recognition (NOR) behavioral assay. Unlike previous therapeutic strategies that primarily focus on inhibiting Tau phosphorylation or directly clearing aggregates, this study highlights the novel role of ANKHD1 in promoting autophagy as a means to mitigate Tau pathology. This novel mechanism not only underscores ANKHD1's potential as a unique therapeutic target for tauopathies but also provides new insights into autophagy-based interventions for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaolin Tian
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA70112, USA
| | - Nathan Le
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA70112, USA
| | - Yuhai Zhao
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA70112, USA
| | - Dina Alawamleh
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA70112, USA
- Louisiana State University Health Sciences Center, School of Medicine, New Orleans, LA70112, USA
| | - Andrew Schwartz
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA70112, USA
- LSU Health Shreveport, School of Medicine, 1501 Kings Highway, Shreveport, Louisiana 71103, USA
| | - Lauren Meyer
- Louisiana State University Health Sciences Center, School of Medicine, New Orleans, LA70112, USA
| | - Elizabeth Helm
- Louisiana State University Health Sciences Center, School of Medicine, New Orleans, LA70112, USA
| | - Chunlai Wu
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA70112, USA
| |
Collapse
|
2
|
Jiang W, Luo H, Zhao M, Fan Q, Ye C, Li X, He J, Lai J, He S, Chen W, Xian W, Chen S, Chen Z, Li D, Chen R, Wang B. Evaluation of canine adipose-derived mesenchymal stem cells for neurological functional recovery in a rat model of traumatic brain injury. BMC Vet Res 2024; 20:110. [PMID: 38500105 PMCID: PMC10946090 DOI: 10.1186/s12917-024-03912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/04/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a common condition in veterinary medicine that is difficult to manage.Veterinary regenerative therapy based on adipose mesenchymal stem cells seem to be an effective strategy for the treatment of traumatic brain injury. In this study, we evaluated therapeutic efficacy of canine Adipose-derived mesenchymal stem cells (AD-MSCs)in a rat TBI model, in terms of improved nerve function and anti-neuroinflammation. RESULTS Canine AD-MSCs promoted neural functional recovery, reduced neuronal apoptosis, and inhibited the activation of microglia and astrocytes in TBI rats. According to the results in vivo, we further investigated the regulatory mechanism of AD-MSCs on activated microglia by co-culture in vitro. Finally, we found that canine AD-MSCs promoted their polarization to the M2 phenotype, and inhibited their polarization to the M1 phenotype. What's more, AD-MSCs could reduce the migration, proliferation and Inflammatory cytokines of activated microglia, which is able to inhibit inflammation in the central system. CONCLUSIONS Collectively, the present study demonstrates that transplantation of canine AD-MSCs can promote functional recovery in TBI rats via inhibition of neuronal apoptosis, glial cell activation and central system inflammation, thus providing a theoretical basis for canine AD-MSCs therapy for TBI in veterinary clinic.
Collapse
Affiliation(s)
- Wenkang Jiang
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, China
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Huina Luo
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Mingming Zhao
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Quanbao Fan
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Cailing Ye
- Deja Lab, VetCell Biotechnology Company Limited, Foshan, 528225, China
| | - Xingying Li
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Jing He
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Jianyi Lai
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Shi He
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Wojun Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Weihang Xian
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Shengfeng Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Zhisheng Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Dongsheng Li
- Deja Lab, VetCell Biotechnology Company Limited, Foshan, 528225, China.
| | - Ruiai Chen
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, China.
| | - Bingyun Wang
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
| |
Collapse
|
3
|
Feng L, Sharma A, Wang Z, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Wiklund L, Sharma HS. Co-administration of Nanowired DL-3-n-Butylphthalide (DL-NBP) Together with Mesenchymal Stem Cells, Monoclonal Antibodies to Alpha Synuclein and TDP-43 (TAR DNA-Binding Protein 43) Enhance Superior Neuroprotection in Parkinson's Disease Following Concussive Head Injury. ADVANCES IN NEUROBIOLOGY 2023; 32:97-138. [PMID: 37480460 DOI: 10.1007/978-3-031-32997-5_3] [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: 07/24/2023]
Abstract
dl-3-n-butylphthalide (dl-NBP) is one of the potent antioxidant compounds that induces profound neuroprotection in stroke and traumatic brain injury. Our previous studies show that dl-NBP reduces brain pathology in Parkinson's disease (PD) following its nanowired delivery together with mesenchymal stem cells (MSCs) exacerbated by concussive head injury (CHI). CHI alone elevates alpha synuclein (ASNC) in brain or cerebrospinal fluid (CSF) associated with elevated TAR DNA-binding protein 43 (TDP-43). TDP-43 protein is also responsible for the pathologies of PD. Thus, it is likely that exacerbation of brain pathology in PD following brain injury may be thwarted using nanowired delivery of monoclonal antibodies (mAb) to ASNC and/or TDP-43. In this review, the co-administration of dl-NBP with MSCs and mAb to ASNC and/or TDP-43 using nanowired delivery in PD and CHI-induced brain pathology is discussed based on our own investigations. Our observations show that co-administration of TiO2 nanowired dl-NBP with MSCs and mAb to ASNC with TDP-43 induced superior neuroprotection in CHI induced exacerbation of brain pathology in PD, not reported earlier.
Collapse
Affiliation(s)
- Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Zhenguo Wang
- Shijiazhuang Pharma Group NBP Pharmaceutical Co., Ltd., Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Kot M, Neglur PK, Pietraszewska A, Buzanska L. Boosting Neurogenesis in the Adult Hippocampus Using Antidepressants and Mesenchymal Stem Cells. Cells 2022; 11:cells11203234. [PMID: 36291101 PMCID: PMC9600461 DOI: 10.3390/cells11203234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
The hippocampus is one of the few privileged regions (neural stem cell niche) of the brain, where neural stem cells differentiate into new neurons throughout adulthood. However, dysregulation of hippocampal neurogenesis with aging, injury, depression and neurodegenerative disease leads to debilitating cognitive impacts. These debilitating symptoms deteriorate the quality of life in the afflicted individuals. Impaired hippocampal neurogenesis is especially difficult to rescue with increasing age and neurodegeneration. However, the potential to boost endogenous Wnt signaling by influencing pathway modulators such as receptors, agonists, and antagonists through drug and cell therapy-based interventions offers hope. Restoration and augmentation of hampered Wnt signaling to facilitate increased hippocampal neurogenesis would serve as an endogenous repair mechanism and contribute to hippocampal structural and functional plasticity. This review focuses on the possible interaction between neurogenesis and Wnt signaling under the control of antidepressants and mesenchymal stem cells (MSCs) to overcome debilitating symptoms caused by age, diseases, or environmental factors such as stress. It will also address some current limitations hindering the direct extrapolation of research from animal models to human application, and the technical challenges associated with the MSCs and their cellular products as potential therapeutic solutions.
Collapse
Affiliation(s)
- Marta Kot
- Correspondence: ; Tel.: +48-22-60-86-563
| | | | | | | |
Collapse
|
5
|
Huang L, Liu M, Jiang W, Ding H, Han Y, Wen M, Li Y, Liu X, Zeng H. Bradykinin/bradykinin 1 receptor promotes brain microvascular endothelial cell permeability and proinflammatory cytokine release by downregulating Wnt3a. J Biochem Mol Toxicol 2022; 36:e23213. [PMID: 36111657 PMCID: PMC10078380 DOI: 10.1002/jbt.23213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/29/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022]
Abstract
Stroke is a life-threatening disease with limited therapeutic options. Damage to the blood-brain barrier (BBB) is the key pathological feature of ischemic stroke. This study explored the role of the bradykinin (BK)/bradykinin 1 receptor (B1R) and its mechanism of action in the BBB. Human brain microvascular endothelial cells (BMECs) were used to test for cellular responses to BK by using the Cell Counting Kit-8 assay, 5-ethynyl-2'-deoxyuridine staining, enzyme-linked immunosorbent assay, flow cytometry, immunofluorescence, cellular permeability assays, and western blotting to evaluate cell viability, cytokine production, and reactive oxygen species (ROS) levels in vitro. A BBB induced by middle cerebral artery occlusion was used to evaluate BBB injuries, and the role played by BK/B1R in ischemic/reperfusion (I/R) was explored in a rat model. Results showed that BK reduced the viability of BMECs and increased the levels of proinflammatory cytokines (interleukin 6 [IL-6], IL-18, and monocyte chemoattractant protein-1) and ROS. Additionally, cellular permeability was increased by BK treatment, and the expression of tight junction proteins (claudin-5 and occludin) was decreased. Interestingly, Wnt3a expression was inhibited by BK and exogenous Wnt3a restored the effects of BK on BMECs. In an in vivo I/R rat model, knockdown of B1R significantly decreased infarct volume and inflammation in I/R rats. Our results suggest that BK might be a key inducer of BBB injury and B1R knockdown might provide a beneficial effect by upregulating Wnt3a.
Collapse
Affiliation(s)
- Linqiang Huang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Mengting Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Wenqiang Jiang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Hongguang Ding
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Yongli Han
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Miaoyun Wen
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| | - Ya Li
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, School of Medicine South China University of Technology Guangzhou China
| | - Xiaoyu Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
- Clinical Medical Division, The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Hongke Zeng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences Guangzhou Guangdong China
| |
Collapse
|
6
|
Characterization of Ex Vivo and In Vitro Wnt Transcriptome Induced by Spinal Cord Injury in Rat Microglial Cells. Brain Sci 2022; 12:brainsci12060708. [PMID: 35741593 PMCID: PMC9221341 DOI: 10.3390/brainsci12060708] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
It is well known that inflammation is crucial in the onset and progression of neurodegenerative diseases and traumatic central nervous system (CNS) injuries, and that microglia and monocyte-derived macrophages (MDMs) play a pivotal role in neuroinflammation. Therefore, the exploration of molecular signaling pathways that are involved in the microglia/macrophage response might help us to shed light on their eventual therapeutic modulation. Interestingly, there is growing evidence showing that the Wnt family of proteins is involved in different neuropathologies that are characterized by a dysregulated neuroinflammatory response, including spinal cord injury (SCI). Here, we aimed to validate a methodology with competence to assess the physiologically relevant Wnt expression patterns of active microglia and MDMs in a rat model of SCI. For that purpose, we have selected and adapted an in vitro system of primary microglia culture that were stimulated with a lesioned spinal cord extract (SCE), together with an ex vivo protocol of flow cytometry sorting of rat microglia/MDMs at different time-points after contusive SCI. Our study demonstrates that the expression profile of Wnt-related genes in microglia/MDM cells exhibit important differences between these particular scenarios which would be in line with previous studies where similar discrepancies have been described for other molecules. Moreover, our results provide for a first experimental report of the Wnt transcriptome in rat microglia and MDMs after SCI which, together with the research platform that was used in the study, and considering its limitations, we expect might contribute to foster the research on Wnt-driven immunomodulatory therapies.
Collapse
|
7
|
The Effect of Oral Mucosal Mesenchymal Stem Cells on Pathological and Long-Term Outcomes in Experimental Traumatic Brain Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4065118. [PMID: 35528162 PMCID: PMC9071883 DOI: 10.1155/2022/4065118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/26/2022] [Accepted: 04/09/2022] [Indexed: 01/10/2023]
Abstract
Background Neuroprotective effects of stem cells have been shown in some neurologic diseases. In this study, the effect of oral mucosal mesenchymal stem cells (OMSCs) on traumatic brain injury (TBI) was evaluated in long term. Materials and Methods TBI was induced by Marmarou's method. The number of 2 × 106 OMSCs was intravenously injected 1 and 24 h after the injury. Brain edema and pathological outcome were assessed at 24 h and 21 days after the injury. Besides, long-term neurological, motor, and cognitive outcomes were evaluated at days 3, 7, 14, and 21 after the injury. Results OMSCs administration could significantly inhibit microglia proliferation, and reduce brain edema and neuronal damage, at 24 h and 21 days after the injury. Neurological function improvement was observed in the times evaluated in OMSCs group. Cognitive and motor function dysfunction and anxiety-like behavior were prevented especially at 14 and 21 days after the injury in the treatment group. Conclusion According to the results of this study, OMSCs administration after TBI reduced brain edema and neuronal damage, improved neurologic outcome, and prevented memory and motor impairments and anxiety-like behavior in long term.
Collapse
|
8
|
Zhou Z, Ren X, Zheng L, Li A, Zhou W. LncRNA NEAT1 stabilized Wnt3a via U2AF2 and activated Wnt/β-catenin pathway to alleviate ischemia stroke induced injury. Brain Res 2022; 1788:147921. [PMID: 35452660 DOI: 10.1016/j.brainres.2022.147921] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ischaemic stroke is the leading cause of mortality and disability in the world. LncRNA NEAT1 has been shown to play an important role in ischaemic injury, but the molecular mechanism remains unclear. METHODS qRT-PCR was used to determine the expression of lncRNA NEAT1 in OGD/R-induced BV-2 cells. Cell viability was assessed by an MTT assay, and cell apoptosis was assessed by flow cytometry. The expression of related proteins was evaluated by Western blotting and ELISA. The interactions among lncRNA NEAT1, U2AF2 and Wnt3a mRNA was demonstrated by RIP and RNA pulldown assays. XAV-939 was used as an inhibitor of the Wnt/β-catenin pathway. RESULTS LncRNA NEAT1 was found to be downregulated in OGD/R-induced BV-2 cells. Overexpression of lncRNA NEAT1 protected BV-2 cells against OGD/R-induced injury. LncRNA NEAT1 enhanced the stability of Wnt3a mRNA via U2AF2. Knockdown of Wnt3a or blockade of the Wnt/β-catenin pathway rescued the effect of lncRNA NEAT1. CONCLUSIONS LncRNA NEAT1 protected cells against OGD/R-induced apoptosis and the inflammatory response by activating the Wnt/β-catenin pathway through upregulation of Wnt3a in a U2AF2-dependent manner. LncRNA NEAT1 could be a promising therapeutic candidate for ischaemic stroke treatment in the future.
Collapse
Affiliation(s)
- Zhiwen Zhou
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Xiang Ren
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Lijun Zheng
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Aiping Li
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China
| | - Wensheng Zhou
- Departments of Neurology, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, Hunan Province, PR China.
| |
Collapse
|
9
|
Yin Y, Tang L, Liu K, Ding X, Wang D, Chen L. Bone marrow mesenchymal stem cells may attenuate lipopolysaccharide-induced liver injury via inhibiting the NLRP3 inflammasome and hepatocyte pyroptosis. Curr Stem Cell Res Ther 2022; 17:361-369. [PMID: 35392791 DOI: 10.2174/1574888x17666220407103441] [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: 09/02/2021] [Revised: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The transplantation of bone marrow mesenchymal cells (BMSCs) has been shown to be an effective means of treating sepsis-related organ damage. Pytoptotic cell death, in turn, has recently been identified as a key driver of sepsis-related damage. At present, there are few studies on the effect of BMSC transplantation on pytoptotic cell death. OBJECTIVE We explored the ability of BMSCs to attenuate hepatic damage in a pyroptosis-related manner in a rat model of lipopolysaccharide (LPS)-induced liver injury. METHODS Following injury modeling and BMSC transplantation, we assessed the expression of the NLR family, pyrin domain containing 3 (NLRP3) inflammasome and key downstream pyroptosis-related signaling molecules. RESULTS It was found that BMSC transplantation was sufficient to significantly improve rat survival after LPS injection. Significantly reduced expression of the pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18 in rats that had undergone BMSC transplantation compared to control animals. Notably, this activity was superior to single-agent administration of the NLRP3 inhibitor MCC950. CONCLUSION Our data suggest that BMSC transplantation may alleviate LPS-induced hepatic damage by suppressing the activation of the NLRP3 inflammasome and the induction of pyroptotic cell death.
Collapse
Affiliation(s)
- Yunyu Yin
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Lu Tang
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Kui Liu
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Xuefeng Ding
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Daqing Wang
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Li Chen
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| |
Collapse
|
10
|
Chen C, Xu HH, Liu XY, Zhang YS, Zhong L, Wang YW, Xu L, Wei P, Chen YX, Liu P, Hao CR, Jia XL, Hu N, Wu XY, Gu XS, Chen LQ, Li XH. 3D printed collagen/silk fibroin scaffolds carrying the secretome of human umbilical mesenchymal stem cells ameliorated neurological dysfunction after spinal cord injury in rats. Regen Biomater 2022; 9:rbac014. [PMID: 35480857 PMCID: PMC9036898 DOI: 10.1093/rb/rbac014] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/05/2023] Open
Abstract
Abstract
Although implantation of biomaterials carrying mesenchymal stem cells (MSCs) is considered as a promising strategy for ameliorating neural function after spinal cord injury (SCI), there are still some challenges including poor cell survival rate, tumorigenicity and ethics concerns. The performance of the secretome derived from MSCs was more stable, and its clinical transformation was more operable. Cytokine antibody array demonstrated that the secretome of MSCs contained 79 proteins among the 174 proteins analyzed. 3D printed collagen/silk fibroin scaffolds carrying MSCs secretome improved hindlimb locomotor function according to the BBB scores, the inclined-grid climbing test and electrophysiological analysis. Parallel with locomotor function recovery, 3D printed collagen/silk fibroin scaffolds carrying MSCs secretome could further facilitate nerve fiber regeneration, enhance remyelination and accelerate the establishment of synaptic connections at the injury site compared to 3D printed collagen/silk fibroin scaffolds alone group according to magnetic resonance imaging (MRI), diffusion Tensor imaging (DTI), hematoxylin and eosin (HE) staining, Bielschowsky’s silver staining immunofluorescence staining and transmission electron microscopy (TEM). These results indicated the implantation of 3D printed collagen/silk fibroin scaffolds carrying MSCs secretome might be a potential treatment for SCI.
Collapse
Affiliation(s)
- Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People’s Armed Police Forces, Tianjin, 300162, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People’s Armed Police Forces, Tianjin, 300162, China
| | - Xiao-Yin Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Yu-Sheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Lin Zhong
- Department of Hematology, the first affiliated hospital of Chengdu medical college, Chengdu, Sichuan, 610500, China
| | - You-Wei Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lin Xu
- Medical Psychology Section, Hubei General Hospital of Armed Police Force, Wuhan, Hubei, 430071, China
| | - Pan Wei
- Department of Neurosurgery, The First People's Hospital Of Long Quan yi District, Cheng Du 610000, Si Chuan, China
| | - Ya-Xing Chen
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Peng Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chen-Ru Hao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Li Jia
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Yang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xiao-Song Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
11
|
Abstract
Traumatic injury of the central nervous system (CNS) is a worldwide health problem affecting millions of people. Trauma of the CNS, that is, traumatic brain injury (TBI) and spinal cord injury (SCI), lead to massive and progressive cell loss and axonal degeneration, usually with very limited regeneration. At present, there are no treatments to protect injured CNS tissue or to replace the lost tissue. Stem cells are a cell type that by definition can self-renew and give rise to multiple cell lineages. In recent years, therapies using stem and progenitor cells have shown promising effects in experimental CNS trauma, particularly in the acute-subacute stage, but also in chronic injuries. However, the therapeutic mechanisms by which transplanted cells achieve the structural and/or functional improvements are often not clear. Stem cell therapies for CNS trauma can be categorized into 2 main concepts, transplantation of exogenous neural stem cells and neural progenitor cells and recruitment of endogenous stem and progenitor cells. In this review, focusing on the advances during the last decade, we will discuss the major cell therapies, the pros and cons of these 2 concepts for TBI and SCI, and the treatment strategies we believe will be successful.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Corresponding author: Erik Sundström, Department of Neurobiology, Care Sciences and Society (NVS), BioClinicum J9:20, Karolinska University Hospital, S17164 Solna, Sweden.
| |
Collapse
|
12
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [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: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Chen B, An J, Guo YS, Tang J, Zhao JJ, Zhang R, Yang H. Tetramethylpyrazine induces the release of BDNF from BM-MSCs through activation of the PI3K/AKT/CREB pathway. Cell Biol Int 2021; 45:2429-2442. [PMID: 34374467 DOI: 10.1002/cbin.11687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/27/2022]
Abstract
Compelling evidences suggest that transplantation of bone marrow-derived mesenchymal stem cells (BM-MSCs) can be therapeutically effective for central nervous system (CNS) injuries and neurodegenerative diseases. The therapeutic effect of BM-MSCs mainly attributes to their differentiation into neuron-like cells which replace injured and degenerative neurons. Importantly, the neurotrophic factors released from BM-MSCs can also rescue injured and degenerative neurons, which plays a biologically pivotal role in enhancing neuroregeneration and neurological functional recovery. Tetramethylpyrazine (TMP), the main bioactive ingredient extracted from the traditional Chinese medicinal herb Chuanxiong, has been reported to promote the neuronal differentiation of BM-MSCs. This study aimed to investigate whether TMP regulates the release of neurotrophic factors from BM-MSCs. We examined the effect of TMP on brain-derived neurotrophic factor (BDNF) released from BM-MSCs and elucidated the underlying molecular mechanism. Our results demonstrated that TMP at concentrations of lower than 200 μM increased the release of BDNF in a dose-dependent manner. Furthermore, the effect of TMP on increasing the release of BDNF from BM-MSCs was blocked by inhibiting the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/cAMP-response element binding protein (CREB) pathway. Therefore, we concluded that TMP could induce the release of BDNF from BM-MSCs through activation of the PI3K/AKT/CREB pathway, leading to the formation of neuroprotective and proneurogenic microenvironment. These findings suggest that TMP possesses novel therapeutic potential to promote neuroprotection and neurogenesis through improving the neurotrophic ability of BM-MSCs, which provides a promising nutritional prevention and treatment strategy for CNS injuries and neurodegenerative diseases via the transplantation of TMP-treated BM-MSCs.
Collapse
Affiliation(s)
- Bo Chen
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing An
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yun-Shan Guo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Juan Tang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Jing-Jing Zhao
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hao Yang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Marzano LAS, de Castro FLM, Machado CA, de Barros JLVM, Macedo E Cordeiro T, Simões E Silva AC, Teixeira AL, Silva de Miranda A. Potential Role of Adult Hippocampal Neurogenesis in Traumatic Brain Injury. Curr Med Chem 2021; 29:3392-3419. [PMID: 34561977 DOI: 10.2174/0929867328666210923143713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/28/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
Traumatic brain injury (TBI) is a serious cause of disability and death among young and adult individuals, displaying complex pathophysiology including cellular and molecular mechanisms that are not fully elucidated. Many experimental and clinical studies investigated the potential relationship between TBI and the process by which neurons are formed in the brain, known as neurogenesis. Currently, there are no available treatments for TBI's long-term consequences being the search for novel therapeutic targets, a goal of highest scientific and clinical priority. Some studies evaluated the benefits of treatments aimed at improving neurogenesis in TBI. In this scenario, herein, we reviewed current pre-clinical studies that evaluated different approaches to improving neurogenesis after TBI while achieving better cognitive outcomes, which may consist in interesting approaches for future treatments.
Collapse
Affiliation(s)
- Lucas Alexandre Santos Marzano
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | | | - Caroline Amaral Machado
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, UFMG, Brazil
| | | | - Thiago Macedo E Cordeiro
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Aline Silva de Miranda
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
15
|
Yan Z, Shi X, Wang H, Si C, Liu Q, Du Y. Neurotrophin-3 Promotes the Neuronal Differentiation of BMSCs and Improves Cognitive Function in a Rat Model of Alzheimer's Disease. Front Cell Neurosci 2021; 15:629356. [PMID: 33642999 PMCID: PMC7902862 DOI: 10.3389/fncel.2021.629356] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 01/21/2021] [Indexed: 11/13/2022] Open
Abstract
Transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) has the potential to be developed into an effective treatment for neurodegenerative diseases such as Alzheimer's disease (AD). However, the therapeutic effects of BMSCs are limited by their low neural differentiation rate. We transfected BMSCs with neurotrophin-3 (NT-3), a neurotrophic factor that promotes neuronal differentiation, and investigated the effects of NT-3 gene overexpression on the differentiation of BMSCs into neurons in vitro and in vivo. We further studied the possible molecular mechanisms. We found that overexpression of NT-3 promoted the differentiation of BMSCs into neurons in vitro and in vivo and improved cognitive function in rats with experimental AD. By contrast, silencing NT-3 inhibited the differentiation of BMSCs and decreased cognitive function in rats with AD. The Wnt/β-catenin signaling pathway was involved in the mechanism by which NT-3 gene modification influenced the neuronal differentiation of BMSCs in vitro and in vivo. Our findings support the prospect of using NT-3-transduced BMSCs for the development of novel therapies for AD.
Collapse
Affiliation(s)
- Zhongrui Yan
- Departments of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Jining No. 1 People's Hospital, Jining, China
| | - Xianjing Shi
- Department of Neurology, Jining No. 1 People's Hospital, Jining, China
| | - Hui Wang
- Department of Neurology, Jining No. 1 People's Hospital, Jining, China
| | - Cuiping Si
- Department of Neurology, Jining No. 1 People's Hospital, Jining, China
| | - Qian Liu
- Department of Neurology, Jining No. 1 People's Hospital, Jining, China
| | - Yifeng Du
- Departments of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Departments of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
16
|
Sun C, Fu J, Qu Z, Jia L, Li D, Zhen J, Wang W. Chronic Intermittent Hypobaric Hypoxia Restores Hippocampus Function and Rescues Cognitive Impairments in Chronic Epileptic Rats via Wnt/β-catenin Signaling. Front Mol Neurosci 2021; 13:617143. [PMID: 33584201 PMCID: PMC7874094 DOI: 10.3389/fnmol.2020.617143] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/22/2020] [Indexed: 01/05/2023] Open
Abstract
Epilepsy is a complex neurological disorder with frequent psychiatric, cognitive, and social comorbidities in addition to recurrent seizures. Cognitive impairment, one of the most common comorbidities, has severe adverse effects on quality of life. Chronic intermittent hypobaric hypoxia (CIHH) has demonstrated neuroprotective efficacy in several neurological disease models. In the present study, we examined the effects of CIHH on cognition and hippocampal function in chronic epileptic rats. CIHH treatment rescued deficits in spatial and object memory, hippocampal neurogenesis, and synaptic plasticity in pilocarpine-treated epileptic rats. The Wnt/β-catenin pathway has been implicated in neural stem cell proliferation and synapse development, and Wnt/β-catenin pathway inhibition effectively blocked the neurogenic effects of CIHH. Our findings indicate that CIHH rescues cognitive deficits in epileptic rats via Wnt/β-catenin pathway activation. This study establishes CIHH and Wnt/β-catenin pathway regulators as potential treatments for epilepsy- induced cognitive impairments.
Collapse
Affiliation(s)
- Can Sun
- Key Laboratory of Neurology of Hebei Province, Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Jian Fu
- Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenzhen Qu
- Key Laboratory of Neurology of Hebei Province, Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lijing Jia
- Key Laboratory of Neurology of Hebei Province, Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dongxiao Li
- Key Laboratory of Neurology of Hebei Province, Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junli Zhen
- Key Laboratory of Neurology of Hebei Province, Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Weiping Wang
- Key Laboratory of Neurology of Hebei Province, Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
17
|
Menet R, Lecordier S, ElAli A. Wnt Pathway: An Emerging Player in Vascular and Traumatic Mediated Brain Injuries. Front Physiol 2020; 11:565667. [PMID: 33071819 PMCID: PMC7530281 DOI: 10.3389/fphys.2020.565667] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
The Wnt pathway, which comprises the canonical and non-canonical pathways, is an evolutionarily conserved mechanism that regulates crucial biological aspects throughout the development and adulthood. Emergence and patterning of the nervous and vascular systems are intimately coordinated, a process in which Wnt pathway plays particularly important roles. In the brain, Wnt ligands activate a cell-specific surface receptor complex to induce intracellular signaling cascades regulating neurogenesis, synaptogenesis, neuronal plasticity, synaptic plasticity, angiogenesis, vascular stabilization, and inflammation. The Wnt pathway is tightly regulated in the adult brain to maintain neurovascular functions. Historically, research in neuroscience has emphasized essentially on investigating the pathway in neurodegenerative disorders. Nonetheless, emerging findings have demonstrated that the pathway is deregulated in vascular- and traumatic-mediated brain injuries. These findings are suggesting that the pathway constitutes a promising target for the development of novel therapeutic protective and restorative interventions. Yet, targeting a complex multifunctional signal transduction pathway remains a major challenge. The review aims to summarize the current knowledge regarding the implication of Wnt pathway in the pathobiology of ischemic and hemorrhagic stroke, as well as traumatic brain injury (TBI). Furthermore, the review will present the strategies used so far to manipulate the pathway for therapeutic purposes as to highlight potential future directions.
Collapse
Affiliation(s)
- Romain Menet
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec - Université Laval, Quebec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
18
|
Diaz MF, Horton PD, Kumar A, Livingston M, Mohammadalipour A, Xue H, Skibber MA, Ewere A, Toledano Furman NE, Aroom KR, Zhang S, Gill BS, Cox CS, Wenzel PL. Injury intensifies T cell mediated graft-versus-host disease in a humanized model of traumatic brain injury. Sci Rep 2020; 10:10729. [PMID: 32612177 PMCID: PMC7330041 DOI: 10.1038/s41598-020-67723-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/10/2020] [Indexed: 12/29/2022] Open
Abstract
The immune system plays critical roles in promoting tissue repair during recovery from neurotrauma but is also responsible for unchecked inflammation that causes neuronal cell death, systemic stress, and lethal immunodepression. Understanding the immune response to neurotrauma is an urgent priority, yet current models of traumatic brain injury (TBI) inadequately recapitulate the human immune response. Here, we report the first description of a humanized model of TBI and show that TBI places significant stress on the bone marrow. Hematopoietic cells of the marrow are regionally decimated, with evidence pointing to exacerbation of underlying graft-versus-host disease (GVHD) linked to presence of human T cells in the marrow. Despite complexities of the humanized mouse, marrow aplasia caused by TBI could be alleviated by cell therapy with human bone marrow mesenchymal stromal cells (MSCs). We conclude that MSCs could be used to ameliorate syndromes triggered by hypercytokinemia in settings of secondary inflammatory stimulus that upset marrow homeostasis such as TBI. More broadly, this study highlights the importance of understanding how underlying immune disorders including immunodepression, autoimmunity, and GVHD might be intensified by injury.
Collapse
Affiliation(s)
- Miguel F Diaz
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Paulina D Horton
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Akshita Kumar
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Megan Livingston
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Amina Mohammadalipour
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Hasen Xue
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Max A Skibber
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Adesuwa Ewere
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,School of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Naama E Toledano Furman
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Kevin R Aroom
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Songlin Zhang
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Brijesh S Gill
- Department of Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Charles S Cox
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Pamela L Wenzel
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA. .,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA. .,Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Schomann T, Iljas JD, Que I, Li Y, Suidgeest E, Cruz LJ, Frijns JHM, Chan A, Löwik CMWG, Huisman MA, Mezzanotte L. Multimodal imaging of hair follicle bulge-derived stem cells in a mouse model of traumatic brain injury. Cell Tissue Res 2020; 381:55-69. [PMID: 32036485 PMCID: PMC7306043 DOI: 10.1007/s00441-020-03173-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 01/20/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a devastating event for which current therapies are limited. Stem cell transplantation may lead to recovery of function via different mechanisms, such as cell replacement through differentiation, stimulation of angiogenesis and support to the microenvironment. Adult hair follicle bulge-derived stem cells (HFBSCs) possess neuronal differentiation capacity, are easy to harvest and are relatively immune-privileged, which makes them potential candidates for autologous stem cell-based therapy. In this study, we apply in vivo multimodal, optical and magnetic resonance imaging techniques to investigate the behavior of mouse HFBSCs in a mouse model of TBI. HFBSCs expressed Luc2 and copGFP and were examined for their differentiation capacity in vitro. Subsequently, transduced HFBSCs, preloaded with ferumoxytol, were transplanted next to the TBI lesion (cortical region) in nude mice, 2 days after injury. Brains were fixed for immunohistochemistry 58 days after transplantation. Luc2- and copGFP-expressing, ferumoxytol-loaded HFBSCs showed adequate neuronal differentiation potential in vitro. Bioluminescence of the lesioned brain revealed survival of HFBSCs and magnetic resonance imaging identified their localization in the area of transplantation. Immunohistochemistry showed that transplanted cells stained for nestin and neurofilament protein (NF-Pan). Cells also expressed laminin and fibronectin but extracellular matrix masses were not detected. After 58 days, ferumoxytol could be detected in HFBSCs in brain tissue sections. These results show that HFBSCs are able to survive after brain transplantation and suggest that cells may undergo differentiation towards a neuronal cell lineage, which supports their potential use for cell-based therapy for TBI.
Collapse
Affiliation(s)
- Timo Schomann
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Percuros B.V, Leiden, the Netherlands
| | - Juvita D Iljas
- Percuros B.V, Leiden, the Netherlands
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Ivo Que
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Yuedan Li
- Percuros B.V, Leiden, the Netherlands
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan H M Frijns
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Alan Chan
- Percuros B.V, Leiden, the Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Clemens M W G Löwik
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Margriet A Huisman
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Hair Science Institute, Maastricht, the Netherlands
| | - Laura Mezzanotte
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands.
- Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
20
|
Traumatic brain injury and hippocampal neurogenesis: Functional implications. Exp Neurol 2020; 331:113372. [PMID: 32504636 DOI: 10.1016/j.expneurol.2020.113372] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 05/23/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022]
Abstract
In the adult brain, self-renewing radial-glia like (RGL) progenitor cells have been shown to reside in the subventricular zone and the subgranular zone of the hippocampus. A large body of evidence shows that experiences such as learning, enriched environment and stress can alter proliferation and differentiation of RGL progenitor cells. The progenitor cells present in the subgranular zone of the hippocampus divide to give rise to newborn neurons that migrate to the dentate gyrus where they differentiate into adult granule neurons. These newborn neurons have been found to have a unique role in certain types of hippocampus-dependent learning and memory, including goal-directed behaviors that require pattern separation. Experimental traumatic brain injury (TBI) in rodents has been shown to alter hippocampal neurogenesis, including triggering the acute loss of newborn neurons, as well as progenitor cell hyper-proliferation. In this review, we discuss the role of hippocampal neurogenesis in learning and memory. Furthermore, we review evidence for the molecular mechanisms that contribute to newborn neuron loss, as well as increased progenitor cell proliferation after TBI. Finally, we discuss strategies aimed at enhancing neurogenesis after TBI and their possible therapeutic benefits.
Collapse
|
21
|
Das M, Mayilsamy K, Mohapatra SS, Mohapatra S. Mesenchymal stem cell therapy for the treatment of traumatic brain injury: progress and prospects. Rev Neurosci 2020; 30:839-855. [PMID: 31203262 DOI: 10.1515/revneuro-2019-0002] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/05/2019] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of injury-related mortality and morbidity in the USA and around the world. The survivors may suffer from cognitive and memory deficits, vision and hearing loss, movement disorders, and different psychological problems. The primary insult causes neuronal damage and activates astrocytes and microglia which evokes immune responses causing further damage to the brain. Clinical trials of drugs to recover the neuronal loss are not very successful. Regenerative approaches for TBI using mesenchymal stem cells (MSCs) seem promising. Results of preclinical research have shown that transplantation of MSCs reduced secondary neurodegeneration and neuroinflammation, promoted neurogenesis and angiogenesis, and improved functional outcome in the experimental animals. The functional improvement is not necessarily related to cell engraftment; rather, immunomodulation by molecular factors secreted by MSCs is responsible for the beneficial effects of this therapy. However, MSC therapy has a few drawbacks including tumor formation, which can be avoided by the use of MSC-derived exosomes. This review has focused on the research works published in the field of regenerative therapy using MSCs after TBI and its future direction.
Collapse
Affiliation(s)
- Mahasweta Das
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Karthick Mayilsamy
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
22
|
Guo K, Yao X, Wu W, Yu Z, Li Z, Ma Z, Liu D. HIF-1α/SDF-1/CXCR4 axis reduces neuronal apoptosis via enhancing the bone marrow-derived mesenchymal stromal cell migration in rats with traumatic brain injury. Exp Mol Pathol 2020; 114:104416. [PMID: 32165091 DOI: 10.1016/j.yexmp.2020.104416] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/21/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022]
Abstract
Mesenchymal stromal injection is a promising therapy for traumatic brain injury (TBI). The aim of this study was to explore the effects of the HIF-1α/SDF-1/CXCR4 axis on neuron repair in TBI rats through improving the bone marrow-derived mesenchymalstromal cells (BMSCs) migration. TBI rat models were established. The rats were treated with exogenous SDF-1, and then the neuronal apoptosis in TBI rats was measured. BMSCs from rats were collected, and the roles of NF-κB p65 expression in nuclei, overexpression of SDF-1 and HIF-1α, as well as downregulation of CXCR4 in BMSC migration were identified. HIF-1α- and SDF-1- treated BMSCs were transplanted into TBI rats, after which the neuronal apoptosis and activity of the HIF-1α/SDF-1/CXCR4 axis were detected. Consequently, we found SDF-1 elevated the HIF-1α/SDF-1/CXCR4 activity and presented protective roles in TBI rat hippocampal neurons with reduced neuronal apoptosis. SDF-1 promoted BMSC migration in vitro, and co-effects of SDF-1 and HIF-1α showed strong promotion, while CXCR4 inhibition suppressed BMSC migration. BMSC transplantation activated the HIF-1α/SDF-1/CXCR4 axis and reduced neuronal apoptosis in TBI rats. To conclude, our study demonstrated that the HIF-1α/SDF-1/CXCR4 axis could enhance BMSC migration and alleviate neuronal damage and apoptosis in TBI rats. This study provided novel options for TBI therapy.
Collapse
Affiliation(s)
- Kai Guo
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Xinyu Yao
- Department of Anesthesia, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Weijing Wu
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Ziyi Yu
- Department of Intensive Care Unit,Tangshan Worker Hospital, Tangshan 063000, Hebei, PR China
| | - Zhenzhong Li
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Zenglu Ma
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China
| | - Dengxiang Liu
- Department of Radiotherapy, Xingtai People's Hospital, Xingtai 054031, Hebei, PR China.
| |
Collapse
|
23
|
Yang HC, Zhang M, Wu R, Zheng HQ, Zhang LY, Luo J, Li LL, Hu XQ. C-C chemokine receptor type 2-overexpressing exosomes alleviated experimental post-stroke cognitive impairment by enhancing microglia/macrophage M2 polarization. World J Stem Cells 2020; 12:152-167. [PMID: 32184939 PMCID: PMC7062036 DOI: 10.4252/wjsc.v12.i2.152] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/27/2019] [Accepted: 01/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human-derived mesenchymal stromal cells have been shown to improve cognitive function following experimental stroke. The activity of exosomes has been verified to be comparable to the therapeutic effects of mesenchymal stromal cells. However, the effects of exosomes derived from human umbilical cord mesenchymal stem cells (HUC-MSCs) (ExoCtrl) on post-stroke cognitive impairment (PSCI) have rarely been reported. Moreover, whether exosomes derived from C-C chemokine receptor type 2 (CCR2)-overexpressing HUC-MSCs (ExoCCR2) can enhance the therapeutic effects on PSCI and the possible underlying mechanisms have not been studied.
AIM To investigate the effects of ExoCtrl on PSCI and whether ExoCCR2 can enhance therapeutic effects on PSCI.
METHODS Transmission electron microscopy, qNano® particles analyzer, and Western blotting were employed to determine the morphology and CCR2 expression of ExoCtrl or ExoCCR2. ELISA was used to study the binding capacity of exosomes to CC chemokine ligand 2 (CCL2) in vivo. After the intravenous injection of ExoCtrl or ExoCCR2 into experimental rats, the effect of ExoCtrl and ExoCCR2 on PSCI was assessed by Morris water maze. Remyelination and oligodendrogenesis were analyzed by Western blotting and immunofluorescence microscopy. QRT-PCR and immunofluorescence microscopy were conducted to compare the microglia/macrophage polarization. The infiltration and activation of hematogenous macrophages were analyzed by Western blotting and transwell migration analysis.
RESULTS CCR2-overexpressing HUC-MSCs loaded the CCR2 receptor into their exosomes. The morphology and diameter distribution between ExoCtrl and ExoCCR2 showed no significant difference. ExoCCR2 bound significantly to CCL2 but ExoCtrl showed little CCL2 binding. Although both ExoCCR2 and ExoCtrl showed beneficial effects on PSCI, oligodendrogenesis, remyelination, and microglia/macrophage polarization, ExoCCR2 exhibited a significantly superior beneficial effect. We also found that ExoCCR2 could suppress the CCL2-induced macrophage migration and activation in vivo and in vitro, compared with ExoCtrl treated group.
CONCLUSION CCR2 over-expression enhanced the therapeutic effects of exosomes on the experimental PSCI by promoting M2 microglia/macrophage polarization, enhancing oligodendrogenesis and remyelination. These therapeutic effects are likely through suppressing the CCL2-induced hematogenous macrophage migration and activation.
Collapse
Affiliation(s)
- Huai-Chun Yang
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Min Zhang
- Department of Andrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Rui Wu
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Hai-Qing Zheng
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Li-Ying Zhang
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Jing Luo
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Li-Li Li
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Xi-Quan Hu
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
24
|
Chang CY, Liang MZ, Wu CC, Huang PY, Chen HI, Yet SF, Tsai JW, Kao CF, Chen L. WNT3A Promotes Neuronal Regeneration upon Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21041463. [PMID: 32098078 PMCID: PMC7073099 DOI: 10.3390/ijms21041463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/12/2023] Open
Abstract
The treatment of traumatic brain injury (TBI) remains a challenge due to limited knowledge about the mechanisms underlying neuronal regeneration. This current study compared the expression of WNT genes during regeneration of injured cortical neurons. Recombinant WNT3A showed positive effect in promoting neuronal regeneration via in vitro, ex vivo, and in vivo TBI models. Intranasal administration of WNT3A protein to TBI mice increased the number of NeuN+ neurons without affecting GFAP+ glial cells, compared to control mice, as well as retained motor function based on functional behavior analysis. Our findings demonstrated that WNT3A, 8A, 9B, and 10A promote regeneration of injured cortical neurons. Among these WNTs, WNT3A showed the most promising regenerative potential in vivo, ex vivo, and in vitro.
Collapse
Affiliation(s)
- Chu-Yuan Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Ching-Chih Wu
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Pei-Yuan Huang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Hong-I Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan;
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11574, Taiwan
- Correspondence: (C.-F.K.); (L.C.); Tel.: +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (C.-F.K.); (L.C.); Tel.: +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| |
Collapse
|
25
|
Pinho AG, Cibrão JR, Silva NA, Monteiro S, Salgado AJ. Cell Secretome: Basic Insights and Therapeutic Opportunities for CNS Disorders. Pharmaceuticals (Basel) 2020; 13:E31. [PMID: 32093352 PMCID: PMC7169381 DOI: 10.3390/ph13020031] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
Transplantation of stem cells, in particular mesenchymal stem cells (MSCs), stands as a promising therapy for trauma, stroke or neurodegenerative conditions such as spinal cord or traumatic brain injuries (SCI or TBI), ischemic stroke (IS), or Parkinson's disease (PD). Over the last few years, cell transplantation-based approaches have started to focus on the use of cell byproducts, with a strong emphasis on cell secretome. Having this in mind, the present review discusses the current state of the art of secretome-based therapy applications in different central nervous system (CNS) pathologies. For this purpose, the following topics are discussed: (1) What are the main cell secretome sources, composition, and associated collection techniques; (2) Possible differences of the therapeutic potential of the protein and vesicular fraction of the secretome; and (3) Impact of the cell secretome on CNS-related problems such as SCI, TBI, IS, and PD. With this, we aim to clarify some of the main questions that currently exist in the field of secretome-based therapies and consequently gain new knowledge that may help in the clinical application of secretome in CNS disorders.
Collapse
Affiliation(s)
- Andreia G. Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Jorge R. Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nuno A. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (A.G.P.); (J.R.C.); (N.A.S.); (S.M.)
- ICVS/3B’s PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| |
Collapse
|
26
|
Dehghanian F, Soltani Z, Khaksari M. Can Mesenchymal Stem Cells Act Multipotential in Traumatic Brain Injury? J Mol Neurosci 2020; 70:677-688. [PMID: 31897971 DOI: 10.1007/s12031-019-01475-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI), a leading cause of morbidity and mortality throughout the world, will probably become the third cause of death in the world by the year 2020. Lack of effective treatments approved for TBI is a major health problem. TBI is a heterogeneous disease due to the different mechanisms of injury. Therefore, it requires combination therapies or multipotential therapy that can affect multiple targets. In recent years, mesenchymal stem cells (MSCs) transplantation has considered one of the most promising therapeutic strategies to repair of brain injuries including TBI. In these studies, it has been shown that MSCs can migrate to the site of injury and differentiate into the cells secreting growth factors and anti-inflammatory cytokines. The reduction in brain edema, neuroinflammation, microglia accumulation, apoptosis, ischemia, the improvement of motor and cognitive function, and the enhancement in neurogenesis, angiogenesis, and neural stem cells survival, proliferation, and differentiation have been indicated in these studies. However, translation of MSCs research in TBI into a clinical setting will require additional preclinical trials.
Collapse
Affiliation(s)
- Fatemeh Dehghanian
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Bam University of Medical Sciences, Bam, Iran
| | - Zahra Soltani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
27
|
Li A, Zhao J, Fan C, Zhu L, Huang C, Li Q, Gan D, Wen C, Chen M, Lu D. Delivery of exogenous proteins by mesenchymal stem cells attenuates early memory deficits in a murine model of Alzheimer's disease. Neurobiol Aging 2019; 86:81-91. [PMID: 31837910 DOI: 10.1016/j.neurobiolaging.2019.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 01/04/2023]
Abstract
A promising intervention for Alzheimer's disease (AD) would ideally target key pathological factors that are involved in AD pathogenesis. Soluble factors produced by engrafted mesenchymal stem cells (MSCs) mediate potential therapeutic effects in AD. However, these therapeutic benefits are largely hampered by the limited paracrine capacity of MSCs. In this study, we used adenovirus-mediated gene transduction of bone marrow MSCs to deliver exogenous proteins into the brain of APPswe/PSEN1dE9 (APP/PS1) mice in the early stage of impairment. We observed that engrafted MSCs carrying exogenous (C-X3-C motif) ligand 1 (CX3CL1) alone reduced the production of the inflammatory cytokine TNF-ɑ and improved synapse-related protein expression but not cognitive function. Transplantation of MSCs carrying CX3CL1 and Wnt3a (CX3CL1-Wnt3a-MSC) significantly attenuated the learning and memory impairment when compared with a control group. The improvement of neurobehavioral functions in APP/PS1 mice treated with CX3CL1-Wnt3a-MSC was related to the inhibition of microglial neurotoxicity and promotion of hippocampal neurogenesis. Transplantation of CX3CL1-Wnt3a-MSC also regulated phosphoinositide 3-kinase/activated protein kinase B (PI3K/AKT) signaling to inhibit the activity of glycogen synthase kinase 3 beta (GSK3β). Taken together, these results indicate that the delivery of exogenous proteins via MSCs can modulate microglial function and enhance neurogenesis, thereby providing new insights into AD intervention.
Collapse
Affiliation(s)
- An Li
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Jiayi Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Chongzhu Fan
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Lihong Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Cuiqin Huang
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Qin Li
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Danhui Gan
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Caiyan Wen
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China
| | - Mengfei Chen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Daxiang Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
28
|
Muhammad SA. Mesenchymal stromal cell secretome as a therapeutic strategy for traumatic brain injury. Biofactors 2019; 45:880-891. [PMID: 31498511 DOI: 10.1002/biof.1563] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) is a global health problem that is a common cause of disability and mortality. Despite the availability of many treatment options, none is capable of restoring functional and structural recovery of the damaged brain. Both the results of preclinical and clinical studies suggest the use of mesenchymal stromal cells (MSCs) as a therapeutic strategy for structural and functional recovery in TBI. However, recent evidence shows that the neuroprotective potential of MSCs is due to multiple secretions of bioactive molecules that modulate tissue microenvironment for tissue repair and regeneration. The results of preclinical studies indicate the therapeutic benefits of MSC secretome in TBI. Soluble bioactive molecules and extracellular vesicles are the various factors secreted by MSCs that can induce neurogenesis, angiogenesis, neovascularization, and anti-inflammatory activities. This review highlights the neuroprotective effect of MSC secretome for the treatment of TBI. In addition, the possible challenges of secretome as biotherapeutics are identified and how some of the issues raised could be overcome for effective clinical application are also discussed.
Collapse
|
29
|
Liu XY, Wei MG, Liang J, Xu HH, Wang JJ, Wang J, Yang XP, Lv FF, Wang KQ, Duan JH, Tu Y, Zhang S, Chen C, Li XH. Injury-preconditioning secretome of umbilical cord mesenchymal stem cells amplified the neurogenesis and cognitive recovery after severe traumatic brain injury in rats. J Neurochem 2019; 153:230-251. [PMID: 31465551 DOI: 10.1111/jnc.14859] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/10/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a dominant cause of death and permanent disability worldwide. Although TBI could significantly increase the proliferation of adult neural stem cells in the hippocampus, the survival and maturation of newborn cells is markedly low. Increasing evidence suggests that the secretome derived from mesenchymal stem cells (MSCs) would be an ideal alternative to MSC transplantation. The successive and microenvironmentally responsive secretion in MSCs may be critical for the functional benefits provided by transplanted MSCs after TBI. Therefore, it is reasonable to hypothesize that the signaling molecules secreted in response to local tissue damage can further facilitate the therapeutic effect of the MSC secretome. To simulate the complex microenvironment in the injured brain well, we used traumatically injured brain tissue extracts to pretreat umbilical cord mesenchymal stem cells (UCMSCs) in vitro and stereotaxically injected the secretome from traumatic injury-preconditioned UCMSCs into the dentate gyrus of the hippocampus in a rat severe TBI model. The results revealed that compared with the normal secretome, the traumatic injury-preconditioned secretome could significantly further promote the differentiation, migration, and maturation of newborn cells in the dentate gyrus and ultimately improve cognitive function after TBI. Cytokine antibody array suggested that the increased benefits of secretome administration were attributable to the newly produced proteins and up-regulated molecules from the MSC secretome preconditioned by a traumatically injured microenvironment. Our study utilized the traumatic injury-preconditioned secretome to amplify neurogenesis and improve cognitive recovery, suggesting this method may be a novel and safer candidate for nerve repair. Cover Image for this issue: doi: 10.1111/jnc.14741.
Collapse
Affiliation(s)
- Xiao-Yin Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.,Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China.,Tianjin Medical University, Tianjin, China
| | - Meng-Guang Wei
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.,Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Xi-Ping Yang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Fang-Fang Lv
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Ke-Qiang Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Jing-Hao Duan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.,Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.,Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| |
Collapse
|
30
|
Wnt-3a alleviates neuroinflammation after ischemic stroke by modulating the responses of microglia/macrophages and astrocytes. Int Immunopharmacol 2019; 75:105760. [DOI: 10.1016/j.intimp.2019.105760] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
|
31
|
Saha P, Gupta R, Sen T, Sen N. Histone Deacetylase 4 Downregulation Elicits Post-Traumatic Psychiatric Disorders through Impairment of Neurogenesis. J Neurotrauma 2019; 36:3284-3296. [PMID: 31169064 DOI: 10.1089/neu.2019.6373] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
An enduring deficit in neurogenesis largely contributes to the development of severe post-traumatic psychiatric disorders such as anxiety, depression, and memory impairment following traumatic brain injury (TBI); however, the mechanism remains obscure. Here we have shown that an imbalance in the generation of γ-aminobutyric acid (GABA)ergic and glutamatergic neurons due to aberrant induction of vesicular glutamate transporter 1 (vGlut1)-positive glutamatergic cells is responsible for impaired neuronal differentiation in the hippocampus following TBI. To elucidate the molecular mechanism, we found that TBI activates a transcription factor, Pax3, by increasing its acetylation status, and subsequently induces Ngn2 transcription. This event, in turn, augments the vGlut1-expressing glutamatergic neurons and accumulation of excess glutamate in the hippocampus that can affect neuronal differentiation. In our study the acetylation of Pax3 was increased due to loss of its interaction with a deacetylase, histone deacetylase 4 (HDAC4), which was downregulated after TBI. TBI-induced activation of GSK3β was responsible for the degradation of HDAC4. We also showed that overexpression of HDAC4 before TBI reduces Pax3 acetylation by restoring an interaction between HDAC4 and Pax3 in the hippocampus. This event prevents the aberrant induction of vGlut1-positive glutamatergic neurons by decreasing the Ngn2 level and subsequently reinforces the balance between GABAergic and glutamatergic neurons following TBI. Further, we found that overexpression of HDAC4 in the hippocampus improves anxiety, depressive-like behavior, and memory functions following TBI.
Collapse
Affiliation(s)
- Pampa Saha
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rajaneesh Gupta
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tanusree Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Whittaker K, Burgess R, Jones V, Yang Y, Zhou W, Luo S, Wilson J, Huang R. Quantitative proteomic analyses in blood: A window to human health and disease. J Leukoc Biol 2019; 106:759-775. [PMID: 31329329 DOI: 10.1002/jlb.mr1118-440r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/11/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
| | | | | | | | | | - Shuhong Luo
- RayBiotech Life Norcross Georgia USA
- RayBiotech Life Guangzhou Guangdong China
- South China Biochip Research Center Guangzhou Guangdong China
| | | | - Ruo‐Pan Huang
- RayBiotech Life Norcross Georgia USA
- RayBiotech Life Guangzhou Guangdong China
- South China Biochip Research Center Guangzhou Guangdong China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou Medical University Guangzhou China
- Guangdong Provincial Hospital of Chinese Medicine Guangzhou China
| |
Collapse
|
33
|
Long noncoding RNAs interact with mRNAs: A new perspective on the mechanism of premature brain injury. Neurosci Lett 2019; 707:134274. [PMID: 31103728 DOI: 10.1016/j.neulet.2019.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 11/20/2022]
Abstract
The molecular mechanism of premature brain injury induced by inflammation is not fully understood. Long noncoding RNAs (lncRNAs) have been reported to play crucial roles in neurological disorders including brain injury. However, little is known about the regulatory function of lncRNAs in the premature brain. This study investigates differentially expressed lncRNAs and mRNAs as well as their interactions in the premature brain. Lipopolysaccharides (LPS) were used to induce inflammation in premature rodent models. Brain histology was observed via hematoxylin and eosin (HE) staining and CD68 immunostaining. Arraystar microarry was designed for the profiling of differentially expressed lncRNAs and mRNAs in 4 LPS induced premature brains (L group), 4 full-term control brains (C group) and 3 premature brains were not induced by LPS (P group). Bioinformatic analysis was applied to reveal the functions and co-expression relationship of lncRNAs and mRNAs. Three lncRNAs and 2 mRNAs were selected for validation applying quantitative real time polymerase chain reaction (qRT-PCR). This study demonstrates dysregulated lncRNA and mRNA profiles in the premature brains upon inflammatory insult, thus revealing a novel mechanism of premature brain development from a new perspective of the lncRNAs and mRNA coexpression network and providing important insights into the therapy of premature brain injury.
Collapse
|
34
|
Koh EY, Oyeniyi BT, Fox EE, Scerbo M, Tomasek JS, Wade CE, Holcomb JB. Trends in potentially preventable trauma deaths between 2005-2006 and 2012-2013. Am J Surg 2018; 218:501-506. [PMID: 30595330 DOI: 10.1016/j.amjsurg.2018.12.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 10/29/2018] [Accepted: 12/11/2018] [Indexed: 11/15/2022]
Abstract
BACKGROUND Most studies of trauma deaths include non-preventable deaths, potentially limiting successful intervention efforts. In this study we aimed to compare the potentially preventable trauma deaths between 2 time periods at our institution. METHODS Trauma patients who died in our hospital in 2005-2006 or 2012-2013 were included, non-preventable deaths were excluded from analysis. The Mann-Whitney and chi square test were used to compare variables between both time periods. RESULTS 80% of deaths were non-preventable. Between the study time periods there was a decrease in potentially preventable deaths, from 29% to 12%, p < 0.001. Head injury deaths significantly decreased (40.6%-24.6%, p = 0.03), while hemorrhage deaths were stable during both time periods (47.6%-43.1%, p = 0.55). CONCLUSION Potentially preventable trauma deaths decreased during the study period. Hemorrhage remains constant as the leading cause of potentially preventable deaths. Continued research to improve survival from hemorrhage is warranted.
Collapse
Affiliation(s)
- Ezra Y Koh
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA.
| | - Blessing T Oyeniyi
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Erin E Fox
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Michelle Scerbo
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Jeffrey S Tomasek
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA
| | - Charles E Wade
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA
| | - John B Holcomb
- Center for Translational Injury Research, Department of Surgery, McGovern Medical School, University of Texas Health Science Center Houston, Houston, TX, USA
| |
Collapse
|
35
|
Ko HR, Ahn SY, Chang YS, Hwang I, Yun T, Sung DK, Sung SI, Park WS, Ahn JY. Human UCB-MSCs treatment upon intraventricular hemorrhage contributes to attenuate hippocampal neuron loss and circuit damage through BDNF-CREB signaling. Stem Cell Res Ther 2018; 9:326. [PMID: 30463591 PMCID: PMC6249960 DOI: 10.1186/s13287-018-1052-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/15/2022] Open
Abstract
Background Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have been shown to prevent brain damage and improve neurocognition following intraventricular hemorrhage (IVH). However, the molecular mechanisms underlying the effects of hUCB-MSCs are still elusive. Thus, as the hippocampus is essential for learning, memory, and cognitive functions and is intimately involved in the ventricular system, making it a potential site of IVH-induced injury, we determined the molecular basis of the effects of hUCB-derived MSCs on hippocampal neurogenesis and the recovery of hippocampal neural circuits after IVH in a rodent model. Methods We inflicted severe IVH injury on postnatal day 4 (P4) in rats. After confirmation of successful induction of IVH using MRI (P5), intracerebroventricular administration of MSCs (ICV-MSC) was performed at 2 days post-injury (P6). For hippocampal synaptic determination, a rat entorhinal-hippocampus (EH) organotypic slice co-culture (OSC) was performed using day 3 post-IVH brains (P7) with or without ICV-MSCs. A similar strategy of experiments was applied to those rats receiving hUCB-MSC transfected with BDNF-Si-RNA for knockdown of BDNF or scrambled siRNA controls after IVH. The molecular mechanism of the MSCs effects on neurogenesis and the attenuation of neuron death was determined by evaluation of BDNF-TrkB-Akt-CREB signaling axis. Results We showed that treatment with hUCB-MSCs attenuated neuronal loss and promoted neurogenesis in the hippocampus, an area highly vulnerable to IVH-induced brain injury. hUCB-MSCs activate BDNF-TrkB receptor signaling, eliciting intracellular activation of Akt and/or Erk and subsequent phosphorylation of CREB, which is responsible for promoting rat BDNF transcription. In addition to the beneficial effects of neuroprotection and neurogenesis, hUCB-MSCs also contribute to the restoration of impaired synaptic circuits in the hippocampus and improve neurocognitive functions in IVH-injured neonatal rat through BDNF-TrkB-CREB signaling axis activation. Conclusions Our data suggest that hUCB-MSCs possess therapeutic potential for treating neuronal loss and neurocognitive dysfunction in IVH through the activation of intracellular TrkB-CREB signaling that is invoked by hUCB-MSC-secreted BDNF. Electronic supplementary material The online version of this article (10.1186/s13287-018-1052-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Taegwan Yun
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Dong Kyung Sung
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea. .,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea.
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea. .,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea. .,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea. .,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, South Korea.
| |
Collapse
|
36
|
Song P, Xia X, Han T, Fang H, Wang Y, Dong F, Zhang R, Ge P, Shen C. BMSCs promote the differentiation of NSCs into oligodendrocytes via mediating Id2 and Olig expression through BMP/Smad signaling pathway. Biosci Rep 2018; 38:BSR20180303. [PMID: 30143582 PMCID: PMC6147919 DOI: 10.1042/bsr20180303] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 01/01/2023] Open
Abstract
Neural stem cells (NSCs) have emerged as a promising treatment for spinal cord injuries. However, the increasing expression of bone morphogenetic proteins (BMPs) in spinal cord injury lesion sites seems to have contributed to the limited oligodendroglial differentiation and the majority of the astroglial differentiation of NSCs. In the present study, we demonstrate that BMPs promote NSCs differentiation toward astrocytes and prevent them from differentiating into oligodendrocytes. This effect is accompanied by the increasing expression of Id2 and the reduction in Oilg1/2 expression. Treatment with bone marrow stromal cells (BMSCs) can enhance the development of oligodendrocytes in the presence of BMPs. The analysis of Id2, as well as Olig1 and Olig2 gene expression, reveals that the effect of BMPs on these gene expressions is reversed with the addition of BMSCs. In sum, these data strongly suggest that BMSCs can promote the differentiation of NSCs into oligodendrocytes through mediating Id2 and Olig1/2 expression by blocking the BMP/Smad signaling pathway.
Collapse
Affiliation(s)
- Peiwen Song
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Xiang Xia
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Tianyu Han
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Huang Fang
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Ying Wang
- Department of Medical Imaging, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Fulong Dong
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Renjie Zhang
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Peng Ge
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Cailiang Shen
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| |
Collapse
|
37
|
Reza-Zaldivar EE, Hernández-Sapiéns MA, Minjarez B, Gutiérrez-Mercado YK, Márquez-Aguirre AL, Canales-Aguirre AA. Potential Effects of MSC-Derived Exosomes in Neuroplasticity in Alzheimer's Disease. Front Cell Neurosci 2018; 12:317. [PMID: 30319358 PMCID: PMC6165870 DOI: 10.3389/fncel.2018.00317] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia affecting regions of the central nervous system that exhibit synaptic plasticity and are involved in higher brain functions such as learning and memory. AD is characterized by progressive cognitive dysfunction, memory loss and behavioral disturbances of synaptic plasticity and energy metabolism. Cell therapy has emerged as an alternative treatment of AD. The use of adult stem cells, such as neural stem cells and Mesenchymal Stem Cells (MSCs) from bone marrow and adipose tissue, have the potential to decrease cognitive deficits, possibly by reducing neuronal loss through blocking apoptosis, increasing neurogenesis, synaptogenesis and angiogenesis. These processes are mediated primarily by the secretion of many growth factors, anti-inflammatory proteins, membrane receptors, microRNAs (miRNA) and exosomes. Exosomes encapsulate and transfer several functional molecules like proteins, lipids and regulatory RNA which can modify cell metabolism. In the proteomic characterization of the content of MSC-derived exosomes, more than 730 proteins have been identified, some of which are specific cell type markers and others are involved in the regulation of binding and fusion of exosomes with adjacent cells. Furthermore, some factors were found that promote the recruitment, proliferation and differentiation of other cells like neural stem cells. Moreover, within exosomal cargo, a wide range of miRNAs were found, which can control functions related to neural remodeling as well as angiogenic and neurogenic processes. Taking this into consideration, the use of exosomes could be part of a strategy to promote neuroplasticity, improve cognitive impairment and neural replacement in AD. In this review, we describe how exosomes are involved in AD pathology and discuss the therapeutic potential of MSC-derived exosomes mediated by miRNA and protein cargo.
Collapse
Affiliation(s)
- Edwin E Reza-Zaldivar
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Mercedes A Hernández-Sapiéns
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Benito Minjarez
- Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| | - Yanet K Gutiérrez-Mercado
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Ana L Márquez-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Alejandro A Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico.,Profesor del programa de Maestría en Ciencias de la Salud Ambiental, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Guadalajara, Mexico
| |
Collapse
|
38
|
Badner A, Hacker J, Hong J, Mikhail M, Vawda R, Fehlings MG. Splenic involvement in umbilical cord matrix-derived mesenchymal stromal cell-mediated effects following traumatic spinal cord injury. J Neuroinflammation 2018; 15:219. [PMID: 30075797 PMCID: PMC6091078 DOI: 10.1186/s12974-018-1243-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/28/2018] [Indexed: 01/05/2023] Open
Abstract
Background The spleen plays an important role in erythrocyte turnover, adaptive immunity, antibody production, and the mobilization of monocytes/macrophages (Mφ) following tissue injury. In response to trauma, the spleen initiates production of inflammatory cytokines, which in turn recruit immune cells to the inflamed tissue, exacerbating damage. Our previous work has shown that intravenous mesenchymal stromal cell (MSC) infusion has potent immunomodulatory effects following spinal cord injury (SCI), associated with the transplanted cells homing to and persisting within the spleen. Therefore, this work aimed to characterize the relationship between the splenic inflammatory response and SCI pathophysiology, emphasizing splenic involvement in MSC-mediated effects. Methods Using a rodent model of cervical clip-compression SCI, secondary tissue damage and functional recovery were compared between splenectomised rodents and those with a sham procedure. Subsequently, 2.5 million MSCs from the term human umbilical cord matrix cells (HUCMCs) were infused via tail vein at 1-h post-SCI and the effects were assessed in the presence or absence of a spleen. Results Splenectomy alone had no effect on lesion volume, hemorrhage, or inflammation. There was also no significant difference between the groups in functional recovery and those in lesion morphometry. Yet, while the infusion of HUCMCs reduced spinal cord hemorrhage and increased systemic levels of IL-10 in the presence of a spleen, these effects were lost with splenectomy. Further, HUCMC infusion was shown to alter the expression levels of splenic cytokines, suggesting that the spleen is an important target and site of MSC effects. Conclusions Our results provide a link between MSC function and splenic inflammation, a finding that can help tailor the cells/transplantation approach to enhance therapeutic efficacy. Electronic supplementary material The online version of this article (10.1186/s12974-018-1243-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Badner
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Justin Hacker
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario, M5T 2S8, Canada
| | - James Hong
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario, M5T 2S8, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Mirriam Mikhail
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario, M5T 2S8, Canada
| | - Reaz Vawda
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario, M5T 2S8, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Ave, Toronto, Ontario, M5T 2S8, Canada. .,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada. .,Division of Neurosurgery, Toronto Western Hospital, 399 Bathurst St. Suite 4WW-449, Toronto, Ontario, M5T 2S8, Canada.
| |
Collapse
|
39
|
Kuwabara A, Liu J, Kamio Y, Liu A, Lawton MT, Lee JW, Hashimoto T. Protective Effect of Mesenchymal Stem Cells Against the Development of Intracranial Aneurysm Rupture in Mice. Neurosurgery 2018; 81:1021-1028. [PMID: 28431181 DOI: 10.1093/neuros/nyx172] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem or stromal cells found in multiple tissues. Intravenous MSC injections have been used to treat various diseases with an inflammatory component in animals and humans. Inflammation is emerging as a key component of pathophysiology of intracranial aneurysms. Modulation of inflammation by MSCs may affect sustained inflammatory processes that lead to aneurysmal rupture. OBJECTIVE To assess the effect of MSCs on the development of aneurysm rupture using a mouse model. METHODS Intracranial aneurysms were induced with a combination of a single elastase injection into the cerebrospinal fluid and deoxycorticosterone acetate salt-induced hypertension in mice. We administered allogeneic bone marrow-derived MSCs or vehicle, 6 and 9 d after aneurysm induction. RESULTS MSC administration significantly reduced rupture rate (vehicle control vs MSCs, 90% vs 36%; P < .05). In cell culture experiments with an MSC and mast cell coculture, MSCs stabilized mast cells through cyclooxygenase-2 (COX-2)-dependent production of prostaglandin E2, thereby reducing the release of proinflammatory cytokines from mast cells. Pretreatment of MSCs with COX-2 inhibitor, NS-398, abolished the protective effect of MSCs against the development of aneurysm rupture. CONCLUSION Intravenous administration of MSCs after aneurysm formation prevented aneurysmal rupture in mice. The protective effect of MSCs against the development of aneurysm rupture appears to be mediated in part by the stabilization of mast cells by MSCs.
Collapse
Affiliation(s)
- Atsushi Kuwabara
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
| | - Jia Liu
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
| | - Yoshinobu Kamio
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
| | - Airan Liu
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
| | - Michael T Lawton
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
- Departments of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Jae-Woo Lee
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
| | - Tomoki Hashimoto
- Departments of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California
- Departments of Neurological Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
40
|
circFGFR4 Promotes Differentiation of Myoblasts via Binding miR-107 to Relieve Its Inhibition of Wnt3a. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:272-283. [PMID: 29858062 PMCID: PMC5992882 DOI: 10.1016/j.omtn.2018.02.012] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 02/25/2018] [Accepted: 02/27/2018] [Indexed: 12/19/2022]
Abstract
Muscle development is regulated under a series of complicate processes, and non-coding RNAs, such as microRNAs (miRNAs) and circular RNAs (circRNAs), have been reported to play important roles in regulating myoblast proliferation and differentiation. We found that miR-107 expression was high in skeletal muscle of Qinchuan cattle. Overexpression of miR-107 inhibited bovine myoblasts differentiation and protected cells from apoptosis. Wnt3a was identified as a target of miR-107 by luciferase activity, real-time qPCR, and western blotting assays. Knockdown of Wnt3a inhibited bovine myoblasts differentiation and apoptosis, and this effect was similar to miR-107 overexpression. We also found circFGFR4 to promote myoblasts differentiation and to induce cell apoptosis. Via luciferase screening and RNA pull-down assays, circFGFR4 was observed to sponge miR-107. Overexpression of circFGFR4 increased the expression of Wnt3a, whereas this effect was abolished by miR-107. These results demonstrated that circFGFR4 binding miR-107 promotes cell differentiation via targeting Wnt3a in bovine primary myoblasts.
Collapse
|
41
|
Wei ZZ, Zhang JY, Taylor TM, Gu X, Zhao Y, Wei L. Neuroprotective and regenerative roles of intranasal Wnt-3a administration after focal ischemic stroke in mice. J Cereb Blood Flow Metab 2018; 38:404-421. [PMID: 28430000 PMCID: PMC5851145 DOI: 10.1177/0271678x17702669] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 01/31/2023]
Abstract
Wnt signaling is a conserved pathway involved in expansion of neural progenitors and lineage specification during development. However, the role of Wnt signaling in the post-stroke brain has not been well-elucidated. We hypothesized that Wnt-3a would play an important role for neurogenesis and brain repair. Adult male mice were subjected to a focal ischemic stroke targeting the sensorimotor cortex. Mice that received Wnt-3a (2 µg/kg/day, 1 h after stroke and once a day for the next 2 days, intranasal delivery) had reduced infarct volume compared to stroke controls. Wnt-3a intranasal treatment of seven days upregulated the expression of brain-derived growth factor (BDNF), increased the proliferation and migration of neuroblasts from the subventricular zone (SVZ), resulting in increased numbers of newly formed neurons and endothelial cells in the peri-infarct zone. Both the molecular and cellular effects of Wnt-3a were blocked by the Wnt specific inhibitors XAV-939 or Dkk-1. In functional assays, Wnt-3a treatment enhanced the local cerebral blood flow (LCBF) in the peri-infarct, as well as improved sensorimotor functions in a battery of behavioral tests. Together, our data demonstrates that the Wnt-3a signaling can act as a dual neuroprotective and regenerative factor for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zheng Zachory Wei
- Laboratories of Stem Cell Biology and Regenerative Medicine, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - James Ya Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Tammi M Taylor
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yingying Zhao
- Laboratories of Stem Cell Biology and Regenerative Medicine, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ling Wei
- Laboratories of Stem Cell Biology and Regenerative Medicine, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
42
|
Shimizu Y, Ueda Y, Ohshima T. Wnt signaling regulates proliferation and differentiation of radial glia in regenerative processes after stab injury in the optic tectum of adult zebrafish. Glia 2018; 66:1382-1394. [PMID: 29411422 DOI: 10.1002/glia.23311] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 01/03/2023]
Abstract
Zebrafish have superior abilities to generate new neurons in the adult brain and to regenerate brain tissue after brain injury compared with mammals. There exist two types of neural stem cells (NSCs): neuroepithelial-like stem cells (NE) and radial glia (RG) in the optic tectum. We established an optic tectum stab injury model to analyze the function of NSCs in the regenerative condition and confirmed that the injury induced the proliferation of RG, but not NE and that the proliferated RG differentiated into new neurons after the injury. We then analyzed the involvement of Wnt signaling after the injury, using a Wnt reporter line in which canonical Wnt signaling activation induced GFP expression and confirmed that GFP expression was induced specifically in RG after the injury. We also analyzed the expression level of genes related to Wnt signaling, and confirmed that endogenous Wnt antagonist dkk1b expression was significantly decreased after the injury. We observed that Wnt signal inhibitor IWR1 treatment suppressed the proliferation and differentiation of RG after the injury, suggesting that up-regulation of Wnt signaling in RG after the stab injury was required for optic tectum regeneration. We also confirmed that Wnt activation by treatment with GSK3β inhibitor BIO in uninjured zebrafish induced proliferation of RG in the optic tectum. This optic tectum stab injury model is useful for the study of the molecular mechanisms of brain regeneration and analysis of the RG functions in physiological and regenerative conditions.
Collapse
Affiliation(s)
- Yuki Shimizu
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Yuto Ueda
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bio-Science, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| |
Collapse
|
43
|
Li W, Chen R, Lv J, Wang H, Liu Y, Peng Y, Qian Z, Fu G, Nie L. In Vivo Photoacoustic Imaging of Brain Injury and Rehabilitation by High-Efficient Near-Infrared Dye Labeled Mesenchymal Stem Cells with Enhanced Brain Barrier Permeability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700277. [PMID: 29619293 PMCID: PMC5827566 DOI: 10.1002/advs.201700277] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/28/2017] [Indexed: 05/18/2023]
Abstract
Stem cell migration and interaction with pathology are critical to understand the complexity and status of disease recovery progress. However, the dynamic visualization still remains a great challenge due to imaging technical limitation, cell labeling difficulty, or blood-brain barrier (BBB). Herein, fast photoacoustic tomography (PAT) with optical molecular probes is applied to noninvasively monitor traumatic brain injury (TBI) and its rehabilitation. The vascular distribution and TBI hemorrhage are clearly imaged, longitudinally monitored, and quantified. Bone mesenchymal stem cells (BMSCs) labeled with modified Prussian blue particles (PBPs), excellent near-infrared dyes and photoacoustic contrasts, are intravenously injected to the mice for improved observation and efficient therapy. BMSCs are demonstrated to be capable of overcoming BBB with enhanced delivery of PBPs to the brain parenchyma. Notably, the versatile BMSCs are observed by PAT to home to the damage region and repair the ruptured vasculature. Moreover, the wound treated by BMSCs exhibits much faster recovery speed than that without treatment. These findings can potentially provide a new noninvasive and high-resolution approach to image TBI, monitor recovery process, and especially trace BMSCs. This study will stimulate extensive researches on brain diseases and provide promising strategies of dye labeled BMSCs in regenerative medicine.
Collapse
Affiliation(s)
- Weitao Li
- Department of Biomedical EngineeringCollege of Automation EngineeringNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
| | - Ronghe Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102P. R. China
| | - Jing Lv
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102P. R. China
| | - Hongke Wang
- Department of Biomedical EngineeringCollege of Automation EngineeringNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102P. R. China
| | - Yu Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102P. R. China
| | - Ya Peng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102P. R. China
| | - Zhiyu Qian
- Department of Biomedical EngineeringCollege of Automation EngineeringNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology and Innovation Center for Cell Signaling NetworkSchool of Life SciencesXiamen UniversityXiamen361102P. R. China
| | - Liming Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102P. R. China
| |
Collapse
|
44
|
Endoplasmic Reticulum Stress Contributes to the Loss of Newborn Hippocampal Neurons after Traumatic Brain Injury. J Neurosci 2018; 38:2372-2384. [PMID: 29386258 DOI: 10.1523/jneurosci.1756-17.2018] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/17/2017] [Accepted: 01/24/2018] [Indexed: 01/24/2023] Open
Abstract
Adult hippocampal neurogenesis has been shown to be required for certain types of cognitive function. For example, studies have shown that these neurons are critical for pattern separation, the ability to store similar experiences as distinct memories. Although traumatic brain injury (TBI) has been shown to cause the loss of newborn hippocampal neurons, the signaling pathway(s) that triggers their death is unknown. Endoplasmic reticulum (ER) stress activates the PERK-eIF2α pathway that acts to restore ER function and improve cell survival. However, unresolved/intense ER stress activates C/EBP homologous protein (CHOP), leading to cell death. We show that TBI causes the death of hippocampal newborn neurons via CHOP. Using CHOP KO mice, we show that loss of CHOP markedly reduces newborn neuron loss after TBI. Injured CHOP mice performed significantly better in a context fear discrimination task compared with injured wild-type mice. In contrast, the PERK inhibitor GSK2606414 exacerbated doublecortin cell loss and worsened contextual discrimination. Administration of guanabenz (which reduces ER stress) to injured male rats reduced the loss of newborn neurons and improved one-trial contextual fear memory. Interestingly, we also found that the surviving newborn neurons in brain-injured animals had dendritic loss, which was not observed in injured CHOP KO mice or in animals treated with guanabenz. These results indicate that ER stress plays a key role in the death of newborn neurons after TBI. Further, these findings indicate that ER stress can alter dendritic arbors, suggesting a role for ER stress in neuroplasticity and dendritic pathologies.SIGNIFICANCE STATEMENT The hippocampus, a structure in the temporal lobe, is critical for learning and memory. The hippocampus is one of only two areas in which neurons are generated in the adult brain. These newborn neurons are required for certain types of memory, and are particularly vulnerable to traumatic brain injury (TBI). However, the mechanism(s) that causes the loss of these cells after TBI is poorly understood. We show that endoplasmic reticulum (ER) stress pathways are activated in newborn neurons after TBI, and that manipulation of the CHOP cascade improves newborn neuron survival and cognitive outcome. These results suggest that treatments that prevent/resolve ER stress may be beneficial in treating TBI-triggered memory dysfunction.
Collapse
|
45
|
Zhao J, Hylin MJ, Kobori N, Hood KN, Moore AN, Dash PK. Post-Injury Administration of Galantamine Reduces Traumatic Brain Injury Pathology and Improves Outcome. J Neurotrauma 2017; 35:362-374. [PMID: 29088998 DOI: 10.1089/neu.2017.5102] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Acetylcholine is an excitatory neurotransmitter in the central nervous system that plays a key role in cognitive function, including learning and memory. Previous studies have shown that experimental traumatic brain injury (TBI) reduces cholinergic neurotransmission, decreases evoked release of acetylcholine, and alters cholinergic receptor levels. Galantamine (U.S. Food and Drug Administration approved for the treatment of vascular dementia and Alzheimer's disease) has been shown to inhibit acetylcholinesterase activity and allosterically potentiate nicotinic receptor signaling. We investigated whether acute administration of galantamine can reduce TBI pathology and improve cognitive function tested days after the termination of the drug treatment. Post-injury administration of galantamine was found to decrease TBI-triggered blood-brain barrier (BBB) permeability (tested 24 h post-injury), attenuate the loss of both GABAergic and newborn neurons in the ipsilateral hippocampus, and improve hippocampal function (tested 10 days after termination of the drug treatment). Specifically, significant improvements in the Morris water maze, novel object recognition, and context-specific fear memory tasks were observed in injured animals treated with galantamine. Although messenger RNAs for both M1 (Nos2, TLR4, and IL-12ß) and M2 (Arg1, CCL17, and Mcr1) microglial phenotypes were elevated post-TBI, galantamine treatment did not alter microglial polarization tested 24 h and 6 days post-injury. Taken together, these findings support the further investigation of galantamine as a treatment for TBI.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School , Houston, Texas
| | - Michael J Hylin
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School , Houston, Texas
| | - Nobuhide Kobori
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School , Houston, Texas
| | - Kimberly N Hood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School , Houston, Texas
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School , Houston, Texas
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School , Houston, Texas
| |
Collapse
|
46
|
Effects of over-expression of SOD2 in bone marrow-derived mesenchymal stem cells on traumatic brain injury. Cell Tissue Res 2017; 372:67-75. [PMID: 29082445 DOI: 10.1007/s00441-017-2716-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 10/09/2017] [Indexed: 01/09/2023]
Abstract
Intravenous administration of bone marrow-derived mesenchymal stem cells (BM-MSCs) has been shown to promote nerve cell regeneration following traumatic brain injury (TBI). As the anti-oxidant defense systems in neuronal tissue including superoxide dismutase 2 (SOD2) are crucial to defend cell against oxidative stress. We proposed a new stratege to increase the therapeutic effect of MSCs by preventing cells death from oxidative stress. We overexpressed SOD2 in BM-MSCs, transplanted these MSCs into TBI model mice, assessed the protective effect of SOD2 against oxidation-induced apoptosis in BM-MSCs both in vitro and in vivo, evaluated brain functional recovery by the rotarod behavioral test, and tested the oxidation status of TBI mice brain after BM-MSCs transplantation by monitoring the superoxide dismutase, glutathione and malonaldehyde level. We found over-expression of SOD2 protected BM-MSCs from H2O2-induced cell apoptosis. Injection of SOD2 over-expressed BM-MSCs attenuated neuro-inflammation in the ipsilateral cortex of TBI mice, and protected TBI mice against loss of blood-brain barrier integrity. Furthermore, the rotarod behavioral test showed functional recovery of TBI mice after MSC treatment. Our experiments indicated that SOD2-over-expressed BM-MSCs have an improved therapeutic effect on brain injury treatment in TBI mice.
Collapse
|
47
|
Jang S, Cho HH, Park JS, Jeong HS. Non-canonical Wnt mediated neurogenic differentiation of human bone marrow-derived mesenchymal stem cells. Neurosci Lett 2017; 660:68-73. [PMID: 28916299 DOI: 10.1016/j.neulet.2017.09.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs), which are characterized by multipotency and self-renewal, are responsible for tissue regeneration and repair. We have previously reported in adipose tissue-derived MSCs that only Wnt5a is enhanced at neurogenic differentiation, and the mechanism of differentiation is dependent on the Wnt5a/JNK pathway; however, the role of Wnt/MAPK pathway is yet to be investigated in neurogenic differentiation in BM-MSCs. We compared the transcriptional expression of Wnt in neurogenic induced-hBM-MSCs (NI-hBM-MSCs) with that in primary hBM-MSCs, using RT-PCR, qPCR, and western blotting. Although the expression of Wnt1 and Wnt2 was unchanged, the expression of Wnt4, Wnt5a, and Wnt11 increased after neurogenic differentiation. In addition, only the expression of frizzled class receptor (Fzd) 3 gene was increased, but not of most of the Fzds and Wnt ligands in NI-hBM-MSCs. Interestingly, Wnt4, Wnt5a, and Wnt11 gene expressions significantly increased in NI-hBM-MSCs by qPCR. In addition, the protein expression level of Wnt4 and Wnt5a, but not Wnt3, increased after neurogenic induction. Furthermore, the expressions of phosphorylated-GSK-3β, ERK1/2, and PKC decreased; however, JNK was activated after neurogenic differentiation. Thus, non-canonical Wnts, i.e., Wnt4, Wnt5a, and Wnt11, regulate neurogenic differentiation through Fzd3 activation and the increase in downstream targets of JNK, which is one of the non-canonical pathways, in hBM-MSCs.
Collapse
Affiliation(s)
- Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jong-Seong Park
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Gwangju 61469, Republic of Korea; Research Institute of Medical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
48
|
Xu C, Fu F, Li X, Zhang S. Mesenchymal stem cells maintain the microenvironment of central nervous system by regulating the polarization of macrophages/microglia after traumatic brain injury. Int J Neurosci 2017; 127:1124-1135. [PMID: 28464695 DOI: 10.1080/00207454.2017.1325884] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs), which are regarded as promising candidates for cell replacement therapies, are able to regulate immune responses after traumatic brain injury (TBI). Secondary immune response following the mechanical injury is the essential factor leading to the necrosis and apoptosis of neural cells during and after the cerebral edema has subsided and there is lack of efficient agent that can mitigate such neuroinflammation in the clinical application. By means of three molecular pathways (prostaglandin E2 (PGE2), tumor-necrosis-factor-inducible gene 6 protein (TSG-6), and progesterone receptor (PR) and glucocorticoid receptors (GR)), MSCs induce the activation of macrophages/microglia and drive them polarize into the M2 phenotypes, which inhibits the release of pro-inflammatory cytokines and promotes tissue repair and nerve regeneration. The regulation of MSCs and the polarization of macrophages/microglia are dynamically changing based on the inflammatory environment. Under the stimulation of platelet lysate (PL), MSCs also promote the release of pro-inflammatory cytokines. Meanwhile, the statue of macrophages/microglia exerts significant effects on the survival, proliferation, differentiation and activation of MSCs by changing the niche of cells. They form positive feedback loops in maintaining the homeostasis after TBI to relieving the secondary injury and promoting tissue repair. MSC therapies have obtained great achievements in several central nervous system disease clinical trials, which will accelerate the application of MSCs in TBI treatment.
Collapse
Affiliation(s)
- Chao Xu
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Feng Fu
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Xiaohong Li
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Sai Zhang
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| |
Collapse
|
49
|
Jackson ML, Srivastava AK, Cox CS. Preclinical progenitor cell therapy in traumatic brain injury: a meta-analysis. J Surg Res 2017. [PMID: 28624058 DOI: 10.1016/j.jss.2017.02.078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND No treatment is available to reverse injury associated with traumatic brain injury (TBI). Progenitor cell therapies show promise in both preclinical and clinical studies. We conducted a meta-analysis of preclinical studies using progenitor cells to treat TBI. METHODS EMBASE, MEDLINE, Cochrane Review, Biosis, and Google Scholar were searched for articles using prespecified search strategies. Studies meeting inclusion criteria underwent data extraction. Analysis was performed using Review Manager 5.3 according to a fixed-effects model, and all studies underwent quality scoring. RESULTS Of 430 abstracts identified, 38 met inclusion criteria and underwent analysis. Average quality score was 4.32 of 8 possible points. No study achieved a perfect score. Lesion volume (LV) and neurologic severity score (NSS) outcomes favored cell treatment with standard mean difference (SMD) of 0.86 (95% CI: 0.64-1.09) and 1.36 (95% CI: 1.11-1.60), respectively. Rotarod and Morris water maze outcomes also favored treatment with improvements in SMD of 0.34 (95% CI: 0.02-0.65) and 0.46 (95% CI: 0.17-74), respectively. Although LV and NSS were robust to publication bias assessments, rotarod and Morris water maze tests were not. Heterogeneity (I2) ranged from 74%-85% among the analyses, indicating a high amount of heterogeneity among studies. Precision as a function of quality score showed a statistically significant increase in the size of the confidence interval as quality improved. CONCLUSIONS Our meta-analysis study reveals an overall positive effect of progenitor cell therapies on LV and NSS with a trend toward improved motor function and spatial learning in different TBI animal models.
Collapse
Affiliation(s)
- Margaret L Jackson
- Department of Pediatric Surgery, University of Texas Health Sciences Center at Houston, Houston, Texas.
| | - Amit K Srivastava
- Department of Pediatric Surgery, University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Health Sciences Center at Houston, Houston, Texas
| |
Collapse
|
50
|
Mead B, Tomarev S. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. Stem Cells Transl Med 2017; 6:1273-1285. [PMID: 28198592 PMCID: PMC5442835 DOI: 10.1002/sctm.16-0428] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022] Open
Abstract
The loss of retinal ganglion cells (RGC) and their axons is one of the leading causes of blindness and includes traumatic (optic neuropathy) and degenerative (glaucoma) eye diseases. Although no clinical therapies are in use, mesenchymal stem cells (MSC) have demonstrated significant neuroprotective and axogenic effects on RGC in both of the aforementioned models. Recent evidence has shown that MSC secrete exosomes, membrane enclosed vesicles (30–100 nm) containing proteins, mRNA and miRNA which can be delivered to nearby cells. The present study aimed to isolate exosomes from bone marrow‐derived MSC (BMSC) and test them in a rat optic nerve crush (ONC) model. Treatment of primary retinal cultures with BMSC‐exosomes demonstrated significant neuroprotective and neuritogenic effects. Twenty‐one days after ONC and weekly intravitreal exosome injections; optical coherence tomography, electroretinography, and immunohistochemistry was performed. BMSC‐derived exosomes promoted statistically significant survival of RGC and regeneration of their axons while partially preventing RGC axonal loss and RGC dysfunction. Exosomes successfully delivered their cargo into inner retinal layers and the effects were reliant on miRNA, demonstrated by the diminished therapeutic effects of exosomes derived from BMSC after knockdown of Argonaute‐2, a key miRNA effector molecule. This study supports the use of BMSC‐derived exosomes as a cell‐free therapy for traumatic and degenerative ocular disease. Stem Cells Translational Medicine2017;6:1273–1285
Collapse
Affiliation(s)
- Ben Mead
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stanislav Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|