51
|
Intravenous delivery of mesenchymal stem cells protects both white and gray matter in spinal cord ischemia. Brain Res 2020; 1747:147040. [DOI: 10.1016/j.brainres.2020.147040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022]
|
52
|
Askenase PW. COVID-19 therapy with mesenchymal stromal cells (MSC) and convalescent plasma must consider exosome involvement: Do the exosomes in convalescent plasma antagonize the weak immune antibodies? J Extracell Vesicles 2020; 10:e12004. [PMID: 33304473 PMCID: PMC7710130 DOI: 10.1002/jev2.12004] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/12/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Exosome extracellular vesicles as biologic therapy for COVID-19 are discussed for two areas. The first involves the growing use of mesenchymal stromal cells (MSCs) for the profound clinical cytokine storm and severe pneumonia in COVID-19 patients. Instead, it is recommended to treat alternatively with their MSC-released exosomes. This is because many reports in the literature and our data have shown that the release of exosomes from the in vivo administered MSC is actually responsible for their beneficial effects. Further, the exosomes are superior, simpler and clinically more convenient compared to their parental MSC. Additionally, in the context of COVID-19, the known tendency of MSC to intravascularly aggregate causing lung dysfunction might synergize with the pneumonia aspects, and the tendency of MSC peripheral vascular micro aggregates might synergize with the vascular clots of the COVID-19 disease process, causing significant central or peripheral vascular insufficiency. The second exosome therapeutic area for severe COVID-19 involves use of convalescent plasma for its content of acquired immune antibodies that must consider the role in this therapy of contained nearly trillions of exosomes. Many of these derive from activated immune modulating cells and likely can function to transfer miRNAs that acting epigenetically to also influence the convalescent plasma recipient response to the virus. There is sufficient evidence, like recovery of patients with antibody deficiencies, to postulate that the antibodies actually have little effect and that immune resistance is principally due to T cell mechanisms. Further, COVID-19 convalescent plasma has remarkably weak beneficial effects if compared to what was expected from many prior studies. This may be due to the dysfunctional immune response to the infection and resulting weak Ab that may be impaired further by antagonistic exosomes in the convalescent plasma. At the least, pre selection of plasma for the best antibodies and relevant exosomes would produce the most optimum therapy for very severely affected COVID-19 patients.
Collapse
Affiliation(s)
- Philip W. Askenase
- Section of Rheumatology and Clinical ImmunologyDepartment of Internal MedicineYale University School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
53
|
Liang Y, Duan L, Xu X, Li X, Liu M, Chen H, Lu J, Xia J. Mesenchymal Stem Cell-Derived Exosomes for Treatment of Autism Spectrum Disorder. ACS APPLIED BIO MATERIALS 2020; 3:6384-6393. [PMID: 35021769 DOI: 10.1021/acsabm.0c00831] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent breakthroughs in the field of stem cell therapy have brought hope to the treatment of mental diseases. Animal experiments and clinical studies have shown that transplantation of mesenchymal stem cells (MSCs) has a positive effect on the treatment of autism spectrum disorder (ASD). However, the therapeutic efficacy of the MSC transplants was primarily associated with the signals and molecules secreted by the MSCs. Exosomes, for example, the secreted organelles from MSCs, carry bioactive molecules of the MSCs that are essential for the therapeutic effects in ASD treatment. This then inspires us to explore the intranasal delivery of MSC exosomes to brain tissues for the treatment of ASD. Exosomes from human umbilical cord mesenchymal stem cells (hUC-MSCs) that efficiently enter the brain tissue through the intranasal route restore the social ability of the mice and correct the repeated stereotyped behaviors and other abnormal phenotypes in the offspring of valproic acid (VPA)-treated mice, which show autism-like symptoms. The therapeutic efficacy can be attributed at least partially to the anti-inflammatory effect of the MSC exosomes. This work thereby reports brain-specific delivery of hUC-MSC exosomes, as a cell-free therapy to relieve autism-related phenotypes, providing a promising direction for the treatment of mental development disorders.
Collapse
Affiliation(s)
- Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare & Shenzhen Institute of Mental Health, Shenzhen 518003, China.,Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Li Duan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Xiao Xu
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Xingfu Li
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518035, China
| | - Min Liu
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hongfei Chen
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jianping Lu
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare & Shenzhen Institute of Mental Health, Shenzhen 518003, China.,School of Medicine Shenzhen University, Shenzhen 518035, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
54
|
Rocha LA, Gomes ED, Afonso JL, Granja S, Baltazar F, Silva NA, Shoichet MS, Sousa RA, Learmonth DA, Salgado AJ. In vitro Evaluation of ASCs and HUVECs Co-cultures in 3D Biodegradable Hydrogels on Neurite Outgrowth and Vascular Organization. Front Cell Dev Biol 2020; 8:489. [PMID: 32612997 PMCID: PMC7308435 DOI: 10.3389/fcell.2020.00489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular disruption following spinal cord injury (SCI) decisively contributes to the poor functional recovery prognosis facing patients with the condition. Using a previously developed gellan gum hydrogel to which the adhesion motif GRGDS was grafted (GG-GRGDS), this work aimed to understand the ability of adipose-derived stem cells (ASCs) to impact vascular organization of human umbilical vein endothelial cells (HUVECs), and how this in turn affects neurite outgrowth of dorsal root ganglia (DRG) explants. Our data shows that culturing these cells together lead to a synergistic effect as showed by increased stimulation of neuritogenesis on DRG. Importantly, HUVECs were only able to assemble into vascular-like structures when cultured in the presence of ASCs, which shows the capacity of these cells in reorganizing the vascular milieu. Analysis of selected neuroregulatory molecules showed that the co-culture upregulated the secretion of several neurotrophic factors. On the other hand, ASCs, and ASCs + HUVECs presented a similar profile regarding the presence of angiotrophic molecules herein analyzed. Finally, the implantation of GG-GRGDS hydrogels encapsulating ASCs in the chick chorioallantoic membrane (CAM) lead to increases in vascular recruitment toward the hydrogels in comparison to GG-GRGDS alone. This indicates that the combination of ASCs with GG-GRGDS hydrogels could promote re-vascularization in trauma-related injuries in the central nervous system and thus control disease progression and induce functional recovery.
Collapse
Affiliation(s)
- Luís A Rocha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal.,Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - João L Afonso
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Sara Granja
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Fatima Baltazar
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| | - Molly S Shoichet
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rui A Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - David A Learmonth
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Barco, Portugal
| | - Antonio J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimaraes, Portugal
| |
Collapse
|
55
|
Papa S, Pizzetti F, Perale G, Veglianese P, Rossi F. Regenerative medicine for spinal cord injury: focus on stem cells and biomaterials. Expert Opin Biol Ther 2020; 20:1203-1213. [PMID: 32421405 DOI: 10.1080/14712598.2020.1770725] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a dramatic medical pathology consequence of a trauma (primary injury). However, most of the post-traumatic degeneration of the tissue is caused by the so-called secondary injury, which is known to be a multifactorial process. This, indeed, includes a wide spectrum of events: blood-brain barrier dysfunction, local inflammation, neuronal death, demyelination and disconnection of nerve pathways. AREAS COVERED Cell therapy represents a promising cure to target diseases and disorders at the cellular level, by restoring cell population or using cells as carriers of therapeutic cargo. In particular, regenerative medicine with stem cells represents the most appealing category to be used, thanks to their peculiar features. EXPERT OPINION Many preclinical research studies demonstrated that cell treatment can improve animal sensory/motor functions and so demonstrated to be very promising for clinical trials. In particular, recent advances have led to the development of biomaterials aiming to promote in situ cell delivery. This review digs into this topic discussing the possibility of cell treatment to improve medical chances in SCI repair.
Collapse
Affiliation(s)
- Simonetta Papa
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Fabio Pizzetti
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy.,Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI) , Lugano, Switzerland.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology , Vienna, Austria
| | - Pietro Veglianese
- Department of Neuroscience, IRCCS Istituto Di Ricerche Farmacologiche "Mario Negri" , Milan, Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Milan, Italy
| |
Collapse
|
56
|
Wang JL, Ren CH, Feng J, Ou CH, Liu L. Oleanolic acid inhibits mouse spinal cord injury through suppressing inflammation and apoptosis via the blockage of p38 and JNK MAPKs. Biomed Pharmacother 2020; 123:109752. [PMID: 31924596 DOI: 10.1016/j.biopha.2019.109752] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/07/2019] [Accepted: 11/29/2019] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury (SCI) is reported as a devastating disease, leading to tissue loss and neurologic dysfunction. However, there is no effective therapeutic strategy for SCI treatment. Oleanolic acid (OA), as a triterpenoid, has anti-oxidant, anti-inflammatory, and anti-apoptotic activities. However, its regulatory effects on SCI have little to be elucidated, as well as the underlying molecular mechanisms. In this study, we attempted to explore the role of OA in SCI progression. Behavior tests suggested that OA treatments markedly alleviated motor function in SCI mice. Evans blue contents up-regulated in spinal cords of SCI mice were significantly reduced by OA in a dose-dependent manner, demonstrating the improved blood-spinal cord barrier. Moreover, we found that OA treatments significantly reduced the apoptotic cell death in spinal cord samples of SCI mice through decreasing the expression of cleaved Caspase-3. In addition, pro-inflammatory response in SCI mice was significantly attenuated by OA treatments. Furthermore, SCI mice exhibited higher activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) signaling pathways, but these effects were clearly blocked in SCI mice with OA treatments, as evidenced by the down-regulated phosphorylation of p38, c-Jun-NH 2 terminal kinase (JNK), IκB kinase α (IKKα), inhibitor of nuclear factor κB-α (IκBα) and NF-κB. The protective effects of OA against SCI were confirmed in lipopolysaccharide (LPS)-stimulated mouse neurons mainly through the suppression of apoptosis and inflammatory response, which were tightly associated with the blockage of p38 and JNK activation. Together, our data demonstrated that OA treatments could dose-dependently ameliorate spinal cord damage through impeding p38- and JNK-regulated apoptosis and inflammation, and therefore OA might be served as an effective therapeutic agent for SCI treatment.
Collapse
Affiliation(s)
- Jiang-Lin Wang
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Chang-He Ren
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Ce-Hua Ou
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China.
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan Province, 646000, China.
| |
Collapse
|
57
|
Namioka T, Namioka A, Sasaki M, Kataoka-Sasaki Y, Oka S, Nakazaki M, Onodera R, Suzuki J, Sasaki Y, Nagahama H, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells promotes functional recovery in a rat model of chronic cerebral infarction. J Neurosurg 2019; 131:1289-1296. [PMID: 30485210 DOI: 10.3171/2018.5.jns18140] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/10/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Intravenous infusion of mesenchymal stem cells (MSCs) derived from adult bone marrow improves behavioral function in rat models of cerebral infarction. Although clinical studies are ongoing, most studies have focused on the acute or subacute phase of stroke. In the present study, MSCs derived from bone marrow of rats were intravenously infused 8 weeks after the induction of a middle cerebral artery occlusion (MCAO) to investigate whether delayed systemic injection of MSCs improves functional outcome in the chronic phase of stroke in rats. METHODS Eight weeks after induction of the MCAO, the rats were randomized and intravenously infused with either MSCs or vehicle. Ischemic volume and behavioral performance were examined. Blood-brain barrier (BBB) integrity was assessed by quantifying the leakage of Evans blue into the brain parenchyma after intravenous infusion. Immunohistochemical analysis was also performed to evaluate the stability of the BBB. RESULTS Motor recovery was better in the MSC-treated group than in the vehicle-treated group, with rapid improvement (evident at 1 week post-infusion). In MSC-treated rats, reduced BBB leakage and increased microvasculature/repair and neovascularization were observed. CONCLUSIONS These results indicate that the systemic infusion of MSCs results in functional improvement, which is associated with structural changes in the chronic phase of cerebral infarction, including in the stabilization of the BBB.
Collapse
Affiliation(s)
- Takahiro Namioka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ai Namioka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masanori Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- 3Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Yuko Kataoka-Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Oka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahito Nakazaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Rie Onodera
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Junpei Suzuki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuichi Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Nagahama
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jeffery D Kocsis
- 2Department of Neurology, Yale University School of Medicine, New Haven; and
- 3Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Osamu Honmou
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- 3Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
58
|
Namioka A, Namioka T, Sasaki M, Kataoka-Sasaki Y, Oka S, Nakazaki M, Onodera R, Suzuki J, Sasaki Y, Nagahama H, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells for protection against brainstem infarction in a persistent basilar artery occlusion model in the adult rat. J Neurosurg 2019; 131:1308-1316. [PMID: 30485204 DOI: 10.3171/2018.4.jns173121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/05/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Morbidity and mortality in patients with posterior circulation stroke remains an issue despite advances in acute stroke therapies. The intravenous infusion of mesenchymal stem cells (MSCs) elicits therapeutic efficacy in experimental supratentorial stroke models. However, since there are few reliable animal models of ischemia in the posterior circulation, the therapeutic approach with intravenous MSC infusion has not been tested. The objective of this study was to test the hypothesis that intravenously infused MSCs provide functional recovery in a newly developed model of brainstem infarction in rats. METHODS Basilar artery (BA) occlusion (BAO) was established in rats by selectively ligating 4 points of the proximal BA with 10-0 nylon monofilament suture. The intravenous infusion of MSCs was performed 1 day after BAO induction. MRI and histological examinations were performed to assess ischemic lesion volume, while multiple behavioral tests were performed to evaluate functional recovery. RESULTS The MSC-treated group exhibited a greater reduction in ischemic lesion volume, while behavioral testing indicated that the MSC-infused group had greater improvement than the vehicle group 28 days after the MSC infusion. Accumulated infused MSCs were observed in the ischemic brainstem lesion. CONCLUSIONS Infused MSCs may provide neuroprotection to facilitate functional outcomes and reduce ischemic lesion volume as evaluated in a newly developed rat model of persistent BAO.
Collapse
Affiliation(s)
- Ai Namioka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takahiro Namioka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masanori Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- 3Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Yuko Kataoka-Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Oka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahito Nakazaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Rie Onodera
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Junpei Suzuki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuichi Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Nagahama
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jeffery D Kocsis
- 2Department of Neurology, Yale University School of Medicine, New Haven; and
- 3Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Osamu Honmou
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- 3Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
59
|
Kang J, Li Z, Zhi Z, Wang S, Xu G. MiR-21 derived from the exosomes of MSCs regulates the death and differentiation of neurons in patients with spinal cord injury. Gene Ther 2019; 26:491-503. [PMID: 31570818 DOI: 10.1038/s41434-019-0101-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/25/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022]
Abstract
In this study, we aimed to investigate the therapeutic effect of miR-21 in the treatment of spinal cord injury (SCI) as well as its underlying molecular mechanisms. Real-time PCR and western blot were performed to measure the expression of miR-21, PTEN, and PDCD4 in SCI rats. Locomotion recovery assessment, Nissl staining, IHC assay, and TUNEL assay were utilized to observe the therapeutic effect of miR-21 in the treatment of SCI. Bioinformatics analysis and luciferase assay were conducted to establish the signaling pathway of miR-21, PTEN, and PDCD4. The regulatory relationships between miR-21 and PTEN/PDCD4 were further validated by real-time PCR, western blot, MTT assay, and flow cytometry. Compared with sham-operated rats, SCI rats showed decreased expression of miR-21 along with increased expression of PTEN/PDCD4. Exosomes were equally distributed in MSCs transfected with miR-21, PTEN siRNA, or scramble controls. The exosomes isolated from the supernatant of cultured MSCs could improve the functional recovery of SCI rats by reducing SCI-induced neuron loss. In addition, miR-21 was shown to inhibit the expression of PTEN/PDCD4 and suppress neuron cell death. Moreover, PTEN and PDCD4 were validated as virtual targets of miR-21. In addition, the miR-21/PTEN/PDCD4 signaling pathway was shown to enhance cell viability and suppress cell death in vivo. The exosomes collected from the supernatant of transfected MSCs contained miR-21, which could improve the functional recovery of SCI rats and suppress cell death both in vivo and in vitro via the miR-21/PTEN/PDCD4 signaling pathway.
Collapse
Affiliation(s)
- Jian Kang
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
| | - Zhenhuan Li
- Department of Orthopedics, ZhaBei Central Hospital of JingaAn District, Shanghai, 200070, China
| | - Zhongzheng Zhi
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
| | - Shiqiang Wang
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
| | - Guanghui Xu
- Department of Orthopedics, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China.
| |
Collapse
|
60
|
Nakazaki M, Sasaki M, Kataoka-Sasaki Y, Oka S, Suzuki J, Sasaki Y, Nagahama H, Hashi K, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells improves impaired cognitive function in a cerebral small vessel disease model. Neuroscience 2019; 408:361-377. [DOI: 10.1016/j.neuroscience.2019.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/18/2019] [Accepted: 04/07/2019] [Indexed: 12/18/2022]
|
61
|
Picoli CC, Coimbra-Campos LMC, Guerra DAP, Silva WN, Prazeres PHDM, Costa AC, Magno LAV, Romano-Silva MA, Mintz A, Birbrair A. Pericytes Act as Key Players in Spinal Cord Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1327-1337. [PMID: 31014955 DOI: 10.1016/j.ajpath.2019.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 03/08/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Spinal cord injury results in locomotor impairment attributable to the formation of an inhibitory fibrous scar, which prevents axonal regeneration after trauma. The scarcity of knowledge about the molecular and cellular mechanisms involved in scar formation after spinal cord lesion impede the design of effective therapies. Recent studies, by using state-of-the-art technologies, including genetic tracking and blockage of pericytes in combination with optogenetics, reveal that pericyte blockage facilitates axonal regeneration and neuronal integration into the local neural circuitry. Strikingly, a pericyte subset is essential during scarring after spinal cord injury, and its arrest results in motor performance improvement. The arising knowledge from current research will contribute to novel approaches to develop therapies for spinal cord injury. We review novel advances in our understanding of pericyte biology in the spinal cord.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Daniel A P Guerra
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luiz A V Magno
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marco A Romano-Silva
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, New York
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Radiology, Columbia University Medical Center, New York, New York.
| |
Collapse
|
62
|
Hakim R, Covacu R, Zachariadis V, Frostell A, Sankavaram SR, Brundin L, Svensson M. Mesenchymal stem cells transplanted into spinal cord injury adopt immune cell-like characteristics. Stem Cell Res Ther 2019; 10:115. [PMID: 30944028 PMCID: PMC6448247 DOI: 10.1186/s13287-019-1218-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/27/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) and their cellular response to various stimuli have been characterized in great detail in culture conditions. In contrast, the cellular response of MSCs in an in vivo setting is still uncharted territory. In this study, we investigated the cellular response of MSCs following transplantation into spinal cord injury (SCI). Methods Mouse bone marrow-derived MSCs were transplanted 24 h following severe contusion SCI in mice. As controls, MSCs transplanted to the uninjured spinal cord and non-transplanted MSCs were used. At 7 days post transplantation, the MSCs were isolated from the SCI, and their global transcriptional changes, survival, differentiation, proliferation, apoptosis, and phenotypes were investigated using RNA sequencing, immunohistochemistry, and flow cytometry. Results MSCs transplanted into SCI downregulated genes related to cell-cycle regulation/progression, DNA metabolic/biosynthetic process, and DNA repair and upregulated genes related to immune system response, cytokine production/response, response to stress/stimuli, signal transduction and signaling pathways, apoptosis, and phagocytosis/endocytosis. MSCs maintained their surface expression of Sca1 and CD29 but upregulated expression of CD45 following transplantation. Transplanted MSCs maintained their surface expression of MHC-I but upregulated surface expression of MHC-II. Transplanted MSCs survived and proliferated to a low extent, did not express Caspase-3, and did not differentiate into neurons or astrocytes. Conclusion MSCs transplanted into SCI upregulate expression of CD45 and MHC-II and expression of genes related to cytokine production, phagocytosis/endocytosis, and immune cells/response and thereby adopt immune cell-like characteristics within the recipient. Electronic supplementary material The online version of this article (10.1186/s13287-019-1218-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ramil Hakim
- Department of Neurology, Karolinska University Hospital, 17176, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden.,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden
| | - Ruxandra Covacu
- Department of Neurology, Karolinska University Hospital, 17176, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Vasilios Zachariadis
- Department of Oncology and Pathology, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Arvid Frostell
- Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden.,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Sreenivasa Raghavan Sankavaram
- Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Lou Brundin
- Department of Neurology, Karolinska University Hospital, 17176, Stockholm, Sweden. .,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden. .,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden.
| | - Mikael Svensson
- Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden.,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital, 17176, Stockholm, Sweden
| |
Collapse
|
63
|
Vawda R, Badner A, Hong J, Mikhail M, Lakhani A, Dragas R, Xhima K, Barretto T, Librach CL, Fehlings MG. Early Intravenous Infusion of Mesenchymal Stromal Cells Exerts a Tissue Source Age-Dependent Beneficial Effect on Neurovascular Integrity and Neurobehavioral Recovery After Traumatic Cervical Spinal Cord Injury. Stem Cells Transl Med 2019; 8:639-649. [PMID: 30912623 PMCID: PMC6591557 DOI: 10.1002/sctm.18-0192] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 02/04/2019] [Indexed: 12/16/2022] Open
Abstract
Localized vascular disruption after traumatic spinal cord injury (SCI) triggers a cascade of secondary events, including inflammation, gliosis, and scarring, that can further impact recovery. In addition to immunomodulatory and neurotrophic properties, mesenchymal stromal cells (MSCs) possess pericytic characteristics. These features make MSCs an ideal candidate for acute cell therapy targeting vascular disruption, which could reduce the severity of secondary injury, enhance tissue preservation and repair, and ultimately promote functional recovery. A moderately severe cervical clip compression/contusion injury was induced at C7‐T1 in adult female rats, followed by an intravenous tail vein infusion 1 hour post‐SCI of (a) term‐birth human umbilical cord perivascular cells (HUCPVCs); (b) first‐trimester human umbilical cord perivascular cells (FTM HUCPVCs); (c) adult bone marrow mesenchymal stem cells; or (d) vehicle control. Weekly behavioral testing was performed. Rats were sacrificed at 24 hours or 10 weeks post‐SCI and immunohistochemistry and ultrasound imaging were performed. Both term and FTM HUCPVC‐infused rats displayed improved (p < .05) grip strength compared with vehicle controls. However, only FTM HUCPVC‐infusion led to significant weight gain. All cell infusion treatments resulted in reduced glial scarring (p < .05). Cell infusion also led to increased axonal, myelin, and vascular densities (p < .05). Although post‐traumatic cavity volume was reduced with cell infusion, this did not reach significance. Taken together, we demonstrate selective long‐term functional recovery alongside histological improvements with HUCPVC infusion in a clinically relevant model of cervical SCI. Our findings highlight the potential of these cells for acute therapeutic intervention after SCI.
Collapse
Affiliation(s)
- Reaz Vawda
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Anna Badner
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - James Hong
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mirriam Mikhail
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alam Lakhani
- CReATe Fertility Centre, Toronto, Ontario, Canada
| | - Rachel Dragas
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kristiana Xhima
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery and Spinal Program, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
64
|
Trost A, Bruckner D, Rivera FJ, Reitsamer HA. Pericytes in the Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:1-26. [DOI: 10.1007/978-3-030-11093-2_1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
65
|
Mukhamedshina YO, Gracheva OA, Mukhutdinova DM, Chelyshev YA, Rizvanov AA. Mesenchymal stem cells and the neuronal microenvironment in the area of spinal cord injury. Neural Regen Res 2019; 14:227-237. [PMID: 30531002 PMCID: PMC6301181 DOI: 10.4103/1673-5374.244778] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell-based technologies are used as a therapeutic strategy in spinal cord injury (SCI). Mesenchymal stem cells (MSCs), which secrete various neurotrophic factors and cytokines, have immunomodulatory, anti-apoptotic and anti-inflammatory effects, modulate reactivity/phenotype of astrocytes and the microglia, thereby promoting neuroregeneration seem to be the most promising. The therapeutic effect of MSCs is due to a paracrine mechanism of their action, therefore the survival of MSCs and their secretory phenotype is of particular importance. Nevertheless, these data are not always reported in efficacy studies of MSC therapy in SCI. Here, we provide a review with summaries of preclinical trials data evaluating the efficacy of MSCs in animal models of SCI. Based on the data collected, we have tried (1) to establish the behavior of MSCs after transplantation in SCI with an evaluation of cell survival, migration potential, distribution in the area of injured and intact tissue and possible differentiation; (2) to determine the effects MSCs on neuronal microenvironment and correlate them with the efficacy of functional recovery in SCI; (3) to ascertain the conditions under which MSCs demonstrate their best survival and greatest efficacy.
Collapse
Affiliation(s)
- Yana O Mukhamedshina
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University; Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Olga A Gracheva
- Department of Therapy and Clinical Diagnostics with radiology Faculty of Veterinary Medicine, Bauman Kazan State Academy of Veterinary Medicine, Kazan, Russia
| | - Dina M Mukhutdinova
- Department of Therapy and Clinical Diagnostics with radiology Faculty of Veterinary Medicine, Bauman Kazan State Academy of Veterinary Medicine, Kazan, Russia
| | - Yurii A Chelyshev
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert A Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| |
Collapse
|
66
|
Oshigiri T, Sasaki T, Sasaki M, Kataoka-Sasaki Y, Nakazaki M, Oka S, Morita T, Hirota R, Yoshimoto M, Yamashita T, Hashimoto-Torii K, Honmou O. Intravenous Infusion of Mesenchymal Stem Cells Alters Motor Cortex Gene Expression in a Rat Model of Acute Spinal Cord Injury. J Neurotrauma 2018; 36:411-420. [PMID: 29901416 DOI: 10.1089/neu.2018.5793] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Recent evidence has demonstrated that remote responses in the brain, as well as local responses in the injured spinal cord, can be induced after spinal cord injury (SCI). Intravenous infusion of mesenchymal stem cells (MSCs) has been shown to provide functional improvements in SCI through local therapeutic mechanisms that provide neuroprotection, stabilization of the blood-spinal cord barrier, remyelination, and axonal sprouting. In the present study, we examined the brain response that might be associated with the functional improvements induced by the infused MSCs after SCI. Genome-wide RNA profiling was performed in the motor cortex of SCI rats at 3 days post-MSC or vehicle infusion. Then, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) data revealed that the "behaviorally-associated differentially expressed genes (DEGs)" were identified by the Pearson's correlation analysis with the behavioral function, suggesting that the "behaviorally-associated DEGs" may be related to the functional recovery after systemic infusion of MSCs in SCI. These results suggested that the infused MSCs alter the gene expression signature in the brain and that these expression changes may contribute to the improved function in SCI.
Collapse
Affiliation(s)
- Tsutomu Oshigiri
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,2 Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toru Sasaki
- 3 Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Masanori Sasaki
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuko Kataoka-Sasaki
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahito Nakazaki
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Oka
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomonori Morita
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,2 Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryosuke Hirota
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,2 Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsunori Yoshimoto
- 2 Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiko Yamashita
- 2 Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazue Hashimoto-Torii
- 3 Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC.,4 Department of Pediatrics, Pharmacology, and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC.,5 Department of Neurobiology, School of Medicine, Yale University, New Haven, Connecticut
| | - Osamu Honmou
- 1 Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
67
|
Kumar H, Choi H, Jo MJ, Joshi HP, Muttigi M, Bonanomi D, Kim SB, Ban E, Kim A, Lee SH, Kim KT, Sohn S, Zeng X, Han I. Neutrophil elastase inhibition effectively rescued angiopoietin-1 decrease and inhibits glial scar after spinal cord injury. Acta Neuropathol Commun 2018; 6:73. [PMID: 30086801 PMCID: PMC6080383 DOI: 10.1186/s40478-018-0576-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/23/2018] [Indexed: 01/13/2023] Open
Abstract
After spinal cord injury (SCI), neutrophil elastase (NE) released at injury site disrupts vascular endothelium integrity and stabilization. Angiopoietins (ANGPTs) are vascular growth factors that play an important role in vascular stabilization. We hypothesized that neutrophil elastase is one of the key determinants of vascular endothelium disruption/destabilization and affects angiopoietins expression after spinal cord injury. To test this, tubule formation and angiopoietins expression were assessed in endothelial cells exposed to different concentrations of recombinant neutropil elastase. Then, the expression of angiopoietin-1, angiopoietin-2, and neutrophil elastase was determined at 3 h and at 1, 3, 5, 7, 14, 21, and 28 days in a clinically relevant model of moderate compression (35 g for 5 min at T10) spinal cord injury. A dichotomy between the levels of angiopoietin-1 and angiopoietin-2 was observed; thus, we utilized a specific neutrophil elastase inhibitor (sivelestat sodium; 30 mg/kg, i.p., b.i.d.) after spinal cord injury. The expression levels of neutropil elastase and angiopoietin-2 increased, and that of angiopoietin-1 decreased after spinal cord injury in rats. The sivelestat regimen, optimized via a pharmacokinetics study, had potent effects on vascular stabilization by upregulating angiopoietin-1 via the AKT pathway and preventing tight junction protein degradation. Moreover, sivelestat attenuated the levels of inflammatory cytokines and chemokines after spinal cord injury and hence subsequently alleviated secondary damage observed as a reduction in glial scar formation and the promotion of blood vessel formation and stabilization. As a result, hindlimb locomotor function significantly recovered in the sivelestat-treated animals as determined by the Basso, Beattie, and Bresnahan scale and footprint analyses. Furthermore, sivelestat treatment attenuated neuropathic pain as assessed by responses to von Frey filaments after spinal cord injury. Thus, our result suggests that inhibiting neutropil elastase by administration of sivelestat is a promising therapeutic strategy to inhibit glial scar and promote functional recovery by upregulating angiopoietin-1 after spinal cord injury.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Min-Jae Jo
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Hari Prasad Joshi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Manjunatha Muttigi
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Dario Bonanomi
- Molecular Neurobiology Laboratory, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Sung Bum Kim
- Department of Neurosurgery, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Eunmi Ban
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Aeri Kim
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyoung-Tae Kim
- Department of Neurosurgery, Kyungpook National University Hospital, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Neurosurgery, School of Medicine,Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China.
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
68
|
Schultz N, Brännström K, Byman E, Moussaud S, Nielsen HM, Olofsson A, Wennström M. Amyloid-beta 1-40 is associated with alterations in NG2+ pericyte population ex vivo and in vitro. Aging Cell 2018; 17:e12728. [PMID: 29453790 PMCID: PMC5946076 DOI: 10.1111/acel.12728] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/17/2022] Open
Abstract
The population of brain pericytes, a cell type important for vessel stability and blood brain barrier function, has recently been shown altered in patients with Alzheimer's disease (AD). The underlying reason for this alteration is not fully understood, but progressive accumulation of the AD characteristic peptide amyloid‐beta (Aβ) has been suggested as a potential culprit. In the current study, we show reduced number of hippocampal NG2+ pericytes and an association between NG2+ pericyte numbers and Aβ1‐40 levels in AD patients. We further demonstrate, using in vitro studies, an aggregation‐dependent impact of Aβ1‐40 on human NG2+ pericytes. Fibril‐EP Aβ1‐40 exposure reduced pericyte viability and proliferation and increased caspase 3/7 activity. Monomer Aβ1‐40 had quite the opposite effect: increased pericyte viability and proliferation and reduced caspase 3/7 activity. Oligomer‐EP Aβ1‐40 had no impact on either of the cellular events. Our findings add to the growing number of studies suggesting a significant impact on pericytes in the brains of AD patients and suggest different aggregation forms of Aβ1‐40 as potential key regulators of the brain pericyte population size.
Collapse
Affiliation(s)
- Nina Schultz
- Clinical Memory Research Unit; Department of Clinical Sciences Malmö; Lund University; Malmö Sweden
| | | | - Elin Byman
- Clinical Memory Research Unit; Department of Clinical Sciences Malmö; Lund University; Malmö Sweden
| | - Simon Moussaud
- Department of Neurochemistry; Stockholm University; Stockholm Sweden
| | | | - Anders Olofsson
- Department of Medical Biochemistry and Biophysics; Umeå University; Umeå Sweden
| | - Malin Wennström
- Clinical Memory Research Unit; Department of Clinical Sciences Malmö; Lund University; Malmö Sweden
| | | |
Collapse
|
69
|
Lankford KL, Arroyo EJ, Kocsis JD. Postirradiation Necrosis after Slow Microvascular Breakdown in the Adult Rat Spinal Cord is Delayed by Minocycline Treatment. Radiat Res 2018; 190:151-163. [PMID: 29799318 DOI: 10.1667/rr15039.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
To better understand the spatiotemporal course of radiation-induced central nervous system (CNS) vascular necrosis and assess the therapeutic potential of approaches for protecting against radiation-induced necrosis, adult female Sprague Dawley rats received 40 Gy surface dose centered on the T9 thoracic spinal cord segment. Locomotor function, blood-spinal cord barrier (BSCB) integrity and histology were evaluated throughout the study. No functional symptoms were observed for several months postirradiation. However, a sudden onset of paralysis was observed at approximately 5.5 months postirradiation. The progression rapidly led to total paralysis and death within less than 48 h of symptom onset. Open-field locomotor scores and rotarod motor coordination testing showed no evidence of neurological impairment prior to the onset of overt paralysis. Histological examination revealed minimal changes to the vasculature prior to symptom onset. However, Evans blue dye (EvB) extravasation revealed a progressive deterioration of BSCB integrity, beginning at one week postirradiation, affecting regions well outside of the irradiated area. Minocycline treatment significantly delayed the onset of paralysis. The results of this study indicate that extensive asymptomatic disruption of the blood-CNS barrier may precede onset of vascular breakdown by several months and suggests that minocycline treatment has a therapeutic effect by delaying radiation-induced necrosis after CNS irradiation.
Collapse
Affiliation(s)
- Karen L Lankford
- Department of Neurology, Yale University School of Medicine, West Haven, Connecticut
| | - Edgardo J Arroyo
- Center for Neuroscience Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | | |
Collapse
|
70
|
Stamegna JC, Sadelli K, Escoffier G, Girard SD, Veron AD, Bonnet A, Khrestchatisky M, Gauthier P, Roman FS. Grafts of Olfactory Stem Cells Restore Breathing and Motor Functions after Rat Spinal Cord Injury. J Neurotrauma 2018; 35:1765-1780. [PMID: 29357739 DOI: 10.1089/neu.2017.5383] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The transplantation of olfactory ecto-mesenchymal stem cells (OEMSCs) could be a helpful therapeutic strategy for spinal cord repair. Using an acute rat model of high cervical contusion that provokes a persistent hemidiaphragmatic and foreleg paralysis, we evaluated the therapeutic effect of a delayed syngeneic transplantation (two days post-contusion) of OEMSCs within the injured spinal cord. Respiratory function was assessed using diaphragmatic electromyography and neuroelectrophysiological recordings of phrenic nerves (innervating the diaphragm). Locomotor function was evaluated using the ladder-walking locomotor test. Cellular reorganization in the injured area was also studied using immunohistochemical and microscopic techniques. We report a substantial improvement in breathing movements, in activities of the ipsilateral phrenic nerve and ipsilateral diaphragm, and also in locomotor abilities four months post-transplantation with nasal OEMSCs. Moreover, in the grafted spinal cord, axonal disorganization and inflammation were reduced. Some grafted stem cells adopted a neuronal phenotype, and axonal sparing was observed in the injury site. The therapeutic effect on the supraspinal command is presumably because of both neuronal replacements and beneficial paracrine effects on the injury area. Our study provides evidence that nasal OEMSCs could be a first step in clinical application, particularly in patients with reduced breathing/locomotor movements.
Collapse
Affiliation(s)
- Jean-Claude Stamegna
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Kevin Sadelli
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Guy Escoffier
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Stéphane D Girard
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Antoine D Veron
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France .,2 IRSEA, Research Institute in Semiochemistry and Applied Ethology , Apt, France
| | - Amandine Bonnet
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | | | - Patrick Gauthier
- 3 Laboratoire de Neurosciences et Cognitives, Aix-Marseille Université , Marseille, France
| | - François S Roman
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| |
Collapse
|
71
|
Chen NF, Sung CS, Wen ZH, Chen CH, Feng CW, Hung HC, Yang SN, Tsui KH, Chen WF. Therapeutic Effect of Platelet-Rich Plasma in Rat Spinal Cord Injuries. Front Neurosci 2018; 12:252. [PMID: 29740270 PMCID: PMC5924817 DOI: 10.3389/fnins.2018.00252] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/03/2018] [Indexed: 11/13/2022] Open
Abstract
Platelet-rich plasma (PRP) is prepared by centrifuging fresh blood in an anticoagulant state, and harvesting the platelet-rich portion or condensing platelets. Studies have consistently demonstrated that PRP concentrates are an abundant source of growth factors, such as platelet-derived growth factor (PDGF), transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), and epithelial growth factor (EGF). The complex mechanisms underlying spinal cord injury (SCI) diminish intrinsic repair and neuronal regeneration. Several studies have suggested that growth factor-promoted axonal regeneration can occur for an extended period after injury. More importantly, the delivery of exogenous growth factors contained in PRP, such as EGF, IGF-1, and TGF-β, has neurotrophic effects on central nervous system (CNS) injuries and neurodegenerative diseases. However, only a few studies have investigated the effects of PRP on CNS injuries or neurodegenerative diseases. According to our review of relevant literature, no study has investigated the effect of intrathecal (i.t.) PRP injection into the injured spinal cord and activation of intrinsic mechanisms. In the present study, we directly injected i.t. PRP into rat spinal cords and examined the effects of PRP on normal and injured spinal cords. In rats with normal spinal cords, PRP induced microglia and astrocyte activation and PDGF-B and ICAM-1 expression. In rats with SCIs, i.t. PRP enhanced the locomotor recovery and spared white matter, promoted angiogenesis and neuronal regeneration, and modulated blood vessel size. Furthermore, a sustained treatment (a bolus of PRP followed by a 1/3 dose of initial PRP concentration) exerted more favorable therapeutic effects than a single dose of PRP. Our findings suggest by i.t. PRP stimulate angiogenesis, enhancing neuronal regeneration after SCI in rats. Although PRP induces minor inflammation in normal and injured spinal cords, it has many advantages. It is an autologous, biocompatible, nontoxic material that does not result in a major immune response. In addition, based on its safety and ease of preparation, we hypothesize that PRP is a promising therapeutic agent for SCI.
Collapse
Affiliation(s)
- Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Zhi-Hong Wen
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chun-Hong Chen
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - Chien-Wei Feng
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - Han-Chun Hung
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, Taiwan
| | - San-Nan Yang
- School of Medicine, College of Medicine and Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Department of Obstetrics and Gynecology and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurosurgery, Xiamen Chang Gung Hospital, Fujian, China
| |
Collapse
|
72
|
Ham TR, Leipzig ND. Biomaterial strategies for limiting the impact of secondary events following spinal cord injury. Biomed Mater 2018; 13:024105. [PMID: 29155409 PMCID: PMC5824690 DOI: 10.1088/1748-605x/aa9bbb] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The nature of traumatic spinal cord injury (SCI) often involves limited recovery and long-term quality of life complications. The initial injury sets off a variety of secondary cascades, which result in an expanded lesion area. Ultimately, the native tissue fails to regenerate. As treatments are developed in the laboratory, the management of this secondary cascade is an important first step in achieving recovery of normal function. Current literature identifies four broad targets for intervention: inflammation, oxidative stress, disruption of the blood-spinal cord barrier, and formation of an inhibitory glial scar. Because of the complex and interconnected nature of these events, strategies that combine multiple therapies together show much promise. Specifically, approaches that rely on biomaterials to perform a variety of functions are generating intense research interest. In this review, we examine each target and discuss how biomaterials are currently used to address them. Overall, we show that there are an impressive amount of biomaterials and combinatorial treatments which show good promise for slowing secondary events and improving outcomes. If more emphasis is placed on growing our understanding of how materials can manage secondary events, treatments for SCI can be designed in an increasingly rational manner, ultimately improving their potential for translation to the clinic.
Collapse
Affiliation(s)
- Trevor R Ham
- Department of Biomedical Engineering, Auburn Science and Engineering Center 275, West Tower, University of Akron, Akron, OH 44325-3908, United States of America
| | | |
Collapse
|
73
|
Lankford KL, Arroyo EJ, Nazimek K, Bryniarski K, Askenase PW, Kocsis JD. Intravenously delivered mesenchymal stem cell-derived exosomes target M2-type macrophages in the injured spinal cord. PLoS One 2018; 13:e0190358. [PMID: 29293592 PMCID: PMC5749801 DOI: 10.1371/journal.pone.0190358] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 12/13/2017] [Indexed: 02/06/2023] Open
Abstract
In a previous report we showed that intravenous infusion of bone marrow-derived mesenchymal stem cells (MSCs) improved functional recovery after contusive spinal cord injury (SCI) in the non-immunosuppressed rat, although the MSCs themselves were not detected at the spinal cord injury (SCI) site [1]. Rather, the MSCs lodged transiently in the lungs for about two days post-infusion. Preliminary studies and a recent report [2] suggest that the effects of intravenous (IV) infusion of MSCs could be mimicked by IV infusion of exosomes isolated from conditioned media of MSC cultures (MSCexos). In this study, we assessed the possible mechanism of MSCexos action on SCI by investigating the tissue distribution and cellular targeting of DiR fluorescent labeled MSCexos at 3 hours and 24 hours after IV infusion in rats with SCI. The IV delivered MSCexos were detected in contused regions of the spinal cord, but not in the noninjured region of the spinal cord, and were also detected in the spleen, which was notably reduced in weight in the SCI rat, compared to control animals. DiR "hotspots" were specifically associated with CD206-expressing M2 macrophages in the spinal cord and this was confirmed by co-localization with anti-CD63 antibodies labeling a tetraspanin characteristically expressed on exosomes. Our findings that MSCexos specifically target M2-type macrophages at the site of SCI, support the idea that extracellular vesicles, released by MSCs, may mediate at least some of the therapeutic effects of IV MSC administration.
Collapse
Affiliation(s)
- Karen L. Lankford
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Center for Neuroscience Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Edgardo J. Arroyo
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Center for Neuroscience Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University College of Medicine, Krakow; Poland
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University College of Medicine, Krakow; Poland
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Philip W. Askenase
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jeffery D. Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Center for Neuroscience Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, United States of America
| |
Collapse
|
74
|
Zheng B, Zhou Y, Zhang H, Yang G, Hong Z, Han D, Wang Q, He Z, Liu Y, Wu F, Zhang X, Tong S, Xu H, Xiao J. Dl-3-n-butylphthalide prevents the disruption of blood-spinal cord barrier via inhibiting endoplasmic reticulum stress following spinal cord injury. Int J Biol Sci 2017; 13:1520-1531. [PMID: 29230100 PMCID: PMC5723918 DOI: 10.7150/ijbs.21107] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 10/15/2017] [Indexed: 11/25/2022] Open
Abstract
After spinal cord injury (SCI), the destruction of blood-spinal cord barrier (BSCB) is shown to accelerate gathering of noxious blood-derived components in the nervous system, leading to secondary neurodegenerative damages. SCI activates endoplasmic reticulum stress (ER stress), which is considered to evoke secondary damages of neurons and glia. Recent evidence indicates that Dl-3-n-butylphthalide (NBP) has the neuroprotective effect in ischaemic brain injury, but whether it has protective effects on SCI or not is largely unclear. Here, we show that NBP prevented BSCB disruption after SCI via inhibition of ER stress. Following a moderate contusion injury of the T9 level of spinal cord, NBP was administered by oral gavage and further treated once a day. NBP significantly attenuated BSCB permeability and breakdown of adherens junction (AJ) and tight junction (TJ) proteins, then improved locomotion recovery following SCI. The protective role of NBP on BSCB disruption is associated with the restrain of ER stress caused by SCI. Furthermore, NBP considerably constrained the expression of ER stress-associated proteins and degradation of TJ and AJ in human brain microvascular endothelial cells (HBMECs) treated with TG. In conclusion, our results indicate that ER stress is associated with the disruption of BSCB integrity after injury, NBP attenuates BSCB disruption via inhibiting ER stress and improve functional recovery following SCI.
Collapse
Affiliation(s)
- Binbin Zheng
- Department of Orthopaedics, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, 317000 PR China.,Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China.,Molecular Pharmacology Research Center, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Yulong Zhou
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China.,Molecular Pharmacology Research Center, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Hongyu Zhang
- Molecular Pharmacology Research Center, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Guangyong Yang
- Department of Orthopaedics, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, 317000 PR China
| | - Zhenghua Hong
- Department of Orthopaedics, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, 317000 PR China
| | - Dandan Han
- Department of Orthopaedics, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, 317000 PR China
| | - Qingqing Wang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Zili He
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Yanlong Liu
- Molecular Pharmacology Research Center, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Fenzan Wu
- Department of Neurosurgery, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, PR China
| | - Xie Zhang
- Department of Gastroenterology, Ningbo Medical Treatment Center Li Hui-li Hospital, Ningbo, Zhejiang, 315040, PR China
| | - Songlin Tong
- Department of Neurosurgery, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, PR China
| | - Huazi Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, 325035 PR China
| |
Collapse
|
75
|
Lee JY, Choi HY, Park CS, Ju BG, Yune TY. Mithramycin A Improves Functional Recovery by Inhibiting BSCB Disruption and Hemorrhage after Spinal Cord Injury. J Neurotrauma 2017; 35:508-520. [PMID: 29048243 DOI: 10.1089/neu.2017.5235] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
After spinal cord injury (SCI), blood-spinal cord barrier (BSCB) disruption and progressive hemorrhage lead to secondary injury, subsequent apoptosis and/or necrosis of neurons and glia, causing permanent neurological deficits. Growing evidence indicates that mithramycin A (MA), an anti-cancer drug, has neuroprotective effects in ischemic brain injury and Huntington's disease (HD). However, the precise mechanism underlying its protective effects is largely unknown. Here, we examined the effect of MA on BSCB breakdown and hemorrhage as well as subsequent inflammation after SCI. After moderate spinal cord contusion injury at T9, MA (150 μg/kg) was immediately injected intraperitoneally (i.p.) and further injected once a day for 5 days. Our data show that MA attenuated BSCB disruption and hemorrhage, and inhibited the infiltration of neutrophils and macrophages after SCI. Consistent with these findings, the expression of inflammatory mediators was significantly alleviated by MA. MA also inhibited the expression and activation of matrix metalloprotease-9 (MMP-9) after injury, which is known to disrupt BSCB and the degradation of tight junction (TJ) proteins. In addition, the expression of sulfonylurea receptor 1 (SUR1) and transient receptor potential melastatin 4 (TRPM4), which are known to mediate hemorrhage at an early stage after SCI, was significantly blocked by MA treatment. Finally, MA inhibited apoptotic cell death and improved functional recovery after injury. Thus, our results demonstrated that MA improves functional recovery by attenuating BSCB disruption and hemorrhage through the downregulation of SUR1/TRPM4 and MMP-9 after SCI.
Collapse
Affiliation(s)
- Jee Y Lee
- 1 Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| | - Hae Y Choi
- 1 Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| | - Chan S Park
- 2 KHU-KIST Department of Converging Science and Technology, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| | - Bong G Ju
- 3 Department of Life Science, Sogang University , Seoul, Republic of Korea
| | - Tae Y Yune
- 1 Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
- 2 KHU-KIST Department of Converging Science and Technology, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
- 4 Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| |
Collapse
|
76
|
Hachem LD, Ahuja CS, Fehlings MG. Assessment and management of acute spinal cord injury: From point of injury to rehabilitation. J Spinal Cord Med 2017; 40:665-675. [PMID: 28571527 PMCID: PMC5778930 DOI: 10.1080/10790268.2017.1329076] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CONTEXT Spinal cord injury (SCI) is a devastating condition that can lead to significant neurological impairment and reduced quality of life. Despite advancements in our understanding of the pathophysiology and secondary injury mechanisms involved in SCI, there are currently very few effective treatments for this condition. The field, however, is rapidly changing as new treatments are developed and key discoveries are made. METHODS In this review, we outline the pathophysiology, management, and long-term rehabilitation of individuals with traumatic SCI. We also provide an in-depth overview of emerging therapies along the spectrum of the translational pipeline. EVIDENCE SYNTHESIS The concept of "time is spine" refers to the concept which emphasizes the importance of early transfer to specialized centers, early decompressive surgery, and early delivery of other treatments (e.g. blood pressure augmentation, methylprednisolone) to affect long-term outcomes. Another important evolution in management has been the recognition and prevention of the chronic complications of SCI including respiratory compromise, bladder dysfunction, Charcot joints, and pressure sores through directed interventions along with early integration of physical rehabilitation and mobilization. There have also been significant advances in neuroprotective and neuroregenerative strategies for SCI, many of which are actively in clinical trial including riluzole, Cethrin, stem cell transplantation, and the use of functional electrical stimulation. CONCLUSION Pharmacologic treatments, cell-based therapies, and other technology-driven interventions will likely play a combinatorial role in the evolving management of SCI as the field continues to evolve.
Collapse
Affiliation(s)
- Laureen D. Hachem
- Institute of Medical Science, University of Toronto, Toronto, ONT, Canada
| | - Christopher S. Ahuja
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ONT, Canada
- Institute of Medical Science, University of Toronto, Toronto, ONT, Canada
| | - Michael G. Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ONT, Canada
- Institute of Medical Science, University of Toronto, Toronto, ONT, Canada
- McEwen Centre for Regenerative Medicine, UHN, University of Toronto, Toronto, ONT, Canada
- Department of Surgery, University of Toronto, Toronto, ONT, Canada
- Spine Program, University of Toronto, Toronto, ONT, Canada
- McLaughlin Center in Molecular Medicine, University of Toronto, Toronto, ONT, Canada
| |
Collapse
|
77
|
Somkuwar SS, Fannon MJ, Bao Nguyen T, Mandyam CD. Hyper-oligodendrogenesis at the vascular niche and reduced blood-brain barrier integrity in the prefrontal cortex during protracted abstinence. Neuroscience 2017; 362:265-271. [PMID: 28870701 DOI: 10.1016/j.neuroscience.2017.08.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/13/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Alcoholism is a relapsing disorder with limited treatment options, in part due to our limited understanding of the disease etiology. We have recently shown that increased ethanol-seeking in a behavioral model of relapse in a rat model of alcoholism was associated with increased oligodendrogenesis which was positively correlated with platelet/endothelial cell adhesion molecule (PECAM-1) expression in the medial prefrontal cortex (mPFC). The current study investigated whether newly born oligodendrocytes form close physical associations with endothelial cells expressing PECAM-1 and whether these changes were accompanied by altered blood-brain barrier (BBB) integrity. Colableling and confocal analysis demonstrate that newly born oligodendroglia were always located in close physical proximity to PECAM-1 in the mPFC of rats that were ethanol dependent and demonstrated high propensity for relapse. Notably, the endothelial proximity of new oligodendrocytes was associated with reduced expression of endothelial barrier antigen (SMI-71), a marker for BBB integrity. Furthermore, voluntary wheel running during abstinence enhanced SMI-71 expression in endothelial cells, indicating protection against abstinence-induced reduction in BBB integrity. Taken together, these results suggest that ethanol experience and abstinence disrupts homeostasis in the oligo-vascular niche in the mPFC. Reversing these mechanisms may hold the key to reducing propensity for relapse in individuals with moderate to severe alcohol use disorder.
Collapse
Affiliation(s)
| | | | - Tran Bao Nguyen
- Skaggs School or Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA, USA
| | - Chitra D Mandyam
- VA San Diego Healthcare System, San Diego, CA, USA; Skaggs School or Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA, USA; Department of Anesthesiology, University of California San Diego, CA, USA.
| |
Collapse
|
78
|
Vismara I, Papa S, Rossi F, Forloni G, Veglianese P. Current Options for Cell Therapy in Spinal Cord Injury. Trends Mol Med 2017; 23:831-849. [PMID: 28811172 DOI: 10.1016/j.molmed.2017.07.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/13/2017] [Accepted: 07/16/2017] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a complex pathology that evolves after primary acute mechanical injury, causing further damage to the spinal cord tissue that exacerbates clinical outcomes. Based on encouraging results from preclinical experiments, some cell treatments being translated into clinical practice demonstrate promising and effective improvement in sensory/motor function. Combinatorial treatments of cell and drug/biological factors have been demonstrated to be more effective than cell treatments alone. Recent advances have led to the development of biomaterials aiming to promote in situ cell delivery for SCI, together with combinatorial strategies using drugs/biomolecules to achieve a maximized multitarget approach. This review provides an overview of single and combinatorial regenerative cell treatments as well as potential delivery options to treat SCI.
Collapse
Affiliation(s)
- Irma Vismara
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy; These authors contributed equally to this work
| | - Simonetta Papa
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy; These authors contributed equally to this work
| | - Filippo Rossi
- Dipartimento di Chimica, Materiali e Ingegneria Chimica 'Giulio Natta', Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy
| | - Gianluigi Forloni
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy
| | - Pietro Veglianese
- Dipartimento di Neuroscienze, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156 Milano, Italy.
| |
Collapse
|
79
|
Pathophysiology, mechanisms and applications of mesenchymal stem cells for the treatment of spinal cord injury. Biomed Pharmacother 2017; 91:693-706. [DOI: 10.1016/j.biopha.2017.04.126] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/25/2017] [Accepted: 04/30/2017] [Indexed: 02/06/2023] Open
|
80
|
Abstract
Spinal cord injury (SCI) represents one of the most complicated and heterogeneous pathological processes of central nervous system (CNS) impairments, which is still beyond functional regeneration. Transplantation of mesenchymal stem cells (MSCs) has been shown to promote the repair of the injured spinal cord tissues in animal models, and therefore, there is much interest in the clinical use of these cells. However, many questions which are essential to improve the therapy effects remain unanswered. For instance, the functional roles and related molecular regulatory mechanisms of MSCs in vivo are not yet completely determined. It is important for transplanted cells to migrate into the injured tissue, to survive and undergo neural differentiation, or to play neural protection roles by various mechanisms after SCI. In this review, we will focus on some of the recent knowledge about the biological behavior and function of MSCs in SCI. Meanwhile, we highlight the function of biomaterials to direct the behavior of MSCs based on our series of work on silk fibroin biomaterials and attempt to emphasize combinational strategies such as tissue engineering for functional improvement of SCI.
Collapse
|
81
|
Trost A, Bruckner D, Kaser-Eichberger A, Motloch K, Bogner B, Runge C, Strohmaier C, Couillard-Despres S, Reitsamer HA, Schroedl F. Lymphatic and vascular markers in an optic nerve crush model in rat. Exp Eye Res 2017; 159:30-39. [PMID: 28315338 DOI: 10.1016/j.exer.2017.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/13/2017] [Accepted: 03/12/2017] [Indexed: 01/23/2023]
Abstract
Only few tissues lack lymphatic supply, such as the CNS or the inner eye. However, if the scleral border is compromised due to trauma or tumor, lymphatics are detected in the eye. Since the situation in the optic nerve (ON), part of the CNS, is not clear, the aim of this study is to screen for the presence of lymphatic markers in the healthy and lesioned ON. Brown Norway rats received an unilateral optic nerve crush (ONC) with defined force, leaving the dura intact. Lesioned ONs and unlesioned contralateral controls were analyzed 7 days (n = 5) and 14 days (n = 5) after ONC, with the following markers: PDGFRb (pericyte), Iba1 (microglia), CD68 (macrophages), RECA (endothelial cell), GFAP (astrocyte) as well as LYVE-1 and podoplanin (PDPN; lymphatic markers). Rat skin sections served as positive controls and confocal microscopy in single optical section mode was used for documentation. In healthy ONs, PDGFRb is detected in vessel-like structures, which are associated to RECA positive structures. Some of these PDGFRb+/RECA+ structures are closely associated with LYVE-1+ cells. Homogenous PDPN-immunoreactivity (IR) was detected in healthy ON without vascular appearance, showing no co-localization with LYVE-1 or PDGFRb but co-localization with GFAP. However, in rat skin controls PDPN-IR was co-localized with LYVE-1 and further with RECA in vessel-like structures. In lesioned ONs, numerous PDGFRb+ cells were detected with network-like appearance in the lesion core. The majority of these PDGFRb+ cells were not associated with RECA-IR, but were immunopositive for Iba1 and CD68. Further, single LYVE-1+ cells were detected here. These LYVE-1+ cells were Iba1-positive but PDPN-negative. PDPN-IR was also clearly absent within the lesion site, while LYVE-1+ and PDPN+ structures were both unaltered outside the lesion. In the lesioned area, PDGFRb+/Iba1+/CD68+ network-like cells without vascular association might represent a subtype of microglia/macrophages, potentially involved in repair and phagocytosis. PDPN was detected in non-lymphatic structures in the healthy ON, co-localizing with GFAP but lacking LYVE-1, therefore most likely representing astrocytes. Both, PDPN and GFAP positive structures are absent in the lesion core. At both time points investigated, no lymphatic structures can be identified in the lesioned ON. However, single markers used to identify lymphatics, detected non-lymphatic structures, highlighting the importance of using a panel of markers to properly identify lymphatic structures.
Collapse
Affiliation(s)
- A Trost
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria.
| | - D Bruckner
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria
| | - A Kaser-Eichberger
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria
| | - K Motloch
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria
| | - B Bogner
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria
| | - C Runge
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria
| | - C Strohmaier
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria
| | - S Couillard-Despres
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria; Institute of Experimental Neuroregeneration, Paracelsus Medical University Salzburg, Austria
| | - H A Reitsamer
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - F Schroedl
- Dept Ophthalmology/ Optometry, Research Program Experimental Ophthalmology, Paracelsus Medical University Salzburg, Austria; Department of Anatomy, Paracelsus Medical University Salzburg, Austria
| |
Collapse
|
82
|
Nakazaki M, Sasaki M, Kataoka-Sasaki Y, Oka S, Namioka T, Namioka A, Onodera R, Suzuki J, Sasaki Y, Nagahama H, Mikami T, Wanibuchi M, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells inhibits intracranial hemorrhage after recombinant tissue plasminogen activator therapy for transient middle cerebral artery occlusion in rats. J Neurosurg 2017; 127:917-926. [PMID: 28059661 DOI: 10.3171/2016.8.jns16240] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Reperfusion therapy with intravenous recombinant tissue plasminogen activator (rtPA) is the standard of care for acute ischemic stroke. However, hemorrhagic complications can result. Intravenous infusion of mesenchymal stem cells (MSCs) reduces stroke volume and improves behavioral function in experimental stroke models. One suggested therapeutic mechanism is inhibition of vascular endothelial dysfunction. The objective of this study was to determine whether MSCs suppress hemorrhagic events after rtPA therapy in the acute phase of transient middle cerebral artery occlusion (tMCAO) in rats. METHODS After induction of tMCAO, 4 groups were studied: 1) normal saline [NS]+vehicle, 2) rtPA+vehicle, 3) NS+MSCs, and 4) rtPA+MSCs. The incidence rate of intracerebral hemorrhage, both hemorrhagic and ischemic volume, and behavioral performance were examined. Matrix metalloproteinase-9 (MMP-9) levels in the brain were assessed with zymography. Quantitative analysis of regional cerebral blood flow (rCBF) was performed to assess hemodynamic change in the ischemic lesion. RESULTS The MSC-treated groups (Groups 3 and 4) experienced a greater reduction in the incidence rate of intracerebral hemorrhage and hemorrhagic volume 1 day after tMCAO even if rtPA was received. The application of rtPA enhanced activation of MMP-9, but MSCs inhibited MMP-9 activation. Behavioral testing indicated that both MSC-infused groups had greater improvement than non-MSC groups had, but rtPA+MSCs provided greater improvement than MSCs alone. The rCBF ratio of rtPA groups (Groups 2 and 4) was similar at 2 hours after reperfusion of tMCAO, but both were greater than that in non-rtPA groups. CONCLUSIONS Infused MSCs may inhibit endothelial dysfunction to suppress hemorrhagic events and facilitate functional outcome. Combined therapy of infused MSCs after rtPA therapy facilitated early behavioral recovery.
Collapse
Affiliation(s)
- Masahito Nakazaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and.,Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Takahiro Namioka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Ai Namioka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Rie Onodera
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Junpei Suzuki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Yuichi Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Hiroshi Nagahama
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Takeshi Mikami
- Department of Neurosurgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and.,Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
83
|
Morita T, Sasaki M, Kataoka-Sasaki Y, Nakazaki M, Nagahama H, Oka S, Oshigiri T, Takebayashi T, Yamashita T, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells promotes functional recovery in a model of chronic spinal cord injury. Neuroscience 2016; 335:221-31. [DOI: 10.1016/j.neuroscience.2016.08.037] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 08/19/2016] [Accepted: 08/20/2016] [Indexed: 12/11/2022]
|
84
|
Hu J, Yu Q, Xie L, Zhu H. Targeting the blood-spinal cord barrier: A therapeutic approach to spinal cord protection against ischemia-reperfusion injury. Life Sci 2016; 158:1-6. [PMID: 27329433 DOI: 10.1016/j.lfs.2016.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Abstract
One of the principal functions of physical barriers between the blood and central nervous system protects system (i.e., blood brain barrier and blood-spinal cord barrier) is the protection from toxic and pathogenic agents in the blood. Disruption of blood-spinal cord barrier (BSCB) plays a key role in spinal cord ischemia-reperfusion injury (SCIRI). Following SCIRI, the permeability of the BSCB increases. Maintaining the integrity of the BSCB alleviates the spinal cord injury after spinal cord ischemia. This review summarizes current knowledge of the structure and function of the BSCB and its changes following SCIRI, as well as the prevention and cure of SCIRI and the role of the BSCB.
Collapse
Affiliation(s)
- Ji Hu
- Department of Anesthesiology, Liyuan Hospital of Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430077, Hubei Province, China.
| | - Qijing Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| | - Lijie Xie
- Department of Anesthesiology, Liyuan Hospital of Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430077, Hubei Province, China
| | - Hongfei Zhu
- Department of Anesthesiology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, Hubei Province, China
| |
Collapse
|
85
|
Badner A, Vawda R, Laliberte A, Hong J, Mikhail M, Jose A, Dragas R, Fehlings M. Early Intravenous Delivery of Human Brain Stromal Cells Modulates Systemic Inflammation and Leads to Vasoprotection in Traumatic Spinal Cord Injury. Stem Cells Transl Med 2016; 5:991-1003. [PMID: 27245367 DOI: 10.5966/sctm.2015-0295] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/07/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED : Spinal cord injury (SCI) is a life-threatening condition with multifaceted complications and limited treatment options. In SCI, the initial physical trauma is closely followed by a series of secondary events, including inflammation and blood spinal cord barrier (BSCB) disruption, which further exacerbate injury. This secondary pathology is partially mediated by the systemic immune response to trauma, in which cytokine production leads to the recruitment/activation of inflammatory cells. Because early intravenous delivery of mesenchymal stromal cells (MSCs) has been shown to mitigate inflammation in various models of neurologic disease, this study aimed to assess these effects in a rat model of SCI (C7-T1, 35-gram clip compression) using human brain-derived stromal cells. Quantitative polymerase chain reaction for a human-specific DNA sequence was used to assess cell biodistribution/clearance and confirmed that only a small proportion (approximately 0.001%-0.002%) of cells are delivered to the spinal cord, with the majority residing in the lung, liver, and spleen. Intriguingly, although cell populations drastically declined in all aforementioned organs, there remained a persistent population in the spleen at 7 days. Furthermore, the cell infusion significantly increased splenic and circulating levels of interleukin-10-a potent anti-inflammatory cytokine. Through this suppression of the systemic inflammatory response, the cells also reduced acute spinal cord BSCB permeability, hemorrhage, and lesion volume. These early effects further translated into enhanced functional recovery and tissue sparing 10 weeks after SCI. This work demonstrates an exciting therapeutic approach whereby a minimally invasive cell-transplantation procedure can effectively reduce secondary damage after SCI through systemic immunomodulation. SIGNIFICANCE Central nervous system pericytes (perivascular stromal cells) have recently gained significant attention within the scientific community. In addition to being recognized as major players in neurotrauma, pericytes have been discovered to share a common origin and potentially function with traditionally defined mesenchymal stromal cells (MSCs). Although there have been several in vitro comparisons, the in vivo therapeutic application of human brain-derived stromal cells has not been previously evaluated. This study demonstrates that these cells not only display a MSC phenotype in vitro but also have similar in vivo immunomodulatory effects after spinal cord injury that are more potent than those of non-central nervous system tissue-derived cells. Therefore, these cells are of great interest for therapeutic use in spinal cord injury.
Collapse
Affiliation(s)
- Anna Badner
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Reaz Vawda
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada
| | - Alex Laliberte
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - James Hong
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Mirriam Mikhail
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada
| | - Alejandro Jose
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada
| | - Rachel Dragas
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Michael Fehlings
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
86
|
Kumar H, Ropper AE, Lee SH, Han I. Propitious Therapeutic Modulators to Prevent Blood-Spinal Cord Barrier Disruption in Spinal Cord Injury. Mol Neurobiol 2016; 54:3578-3590. [PMID: 27194298 DOI: 10.1007/s12035-016-9910-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 05/03/2016] [Indexed: 01/09/2023]
Abstract
The blood-spinal cord barrier (BSCB) is a specialized protective barrier that regulates the movement of molecules between blood vessels and the spinal cord parenchyma. Analogous to the blood-brain barrier (BBB), the BSCB plays a crucial role in maintaining the homeostasis and internal environmental stability of the central nervous system (CNS). After spinal cord injury (SCI), BSCB disruption leads to inflammatory cell invasion such as neutrophils and macrophages, contributing to permanent neurological disability. In this review, we focus on the major proteins mediating the BSCB disruption or BSCB repair after SCI. This review is composed of three parts. Section 1. SCI and the BSCB of the review describes critical events involved in the pathophysiology of SCI and their correlation with BSCB integrity/disruption. Section 2. Major proteins involved in BSCB disruption in SCI focuses on the actions of matrix metalloproteinases (MMPs), tumor necrosis factor alpha (TNF-α), heme oxygenase-1 (HO-1), angiopoietins (Angs), bradykinin, nitric oxide (NO), and endothelins (ETs) in BSCB disruption and repair. Section 3. Therapeutic approaches discusses the major therapeutic compounds utilized to date for the prevention of BSCB disruption in animal model of SCI through modulation of several proteins.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Alexander E Ropper
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| | - Inbo Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
87
|
White SV, Czisch CE, Han MH, Plant CD, Harvey AR, Plant GW. Intravenous Transplantation of Mesenchymal Progenitors Distribute Solely to the Lungs and Improve Outcomes in Cervical Spinal Cord Injury. Stem Cells 2016; 34:1812-25. [DOI: 10.1002/stem.2364] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/16/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Seok Voon White
- Department of Neurosurgery; Stanford Partnership for Spinal Cord Injury and Repair, Stanford University School of Medicine; Stanford California USA
- School of Anatomy, Physiology and Human Biology; The University of Western Australia; Perth Western Australia Australia
| | - Chris E. Czisch
- Department of Neurosurgery; Stanford Partnership for Spinal Cord Injury and Repair, Stanford University School of Medicine; Stanford California USA
| | - May H. Han
- Department of Neurology and Neurological Sciences; Stanford University; Stanford California USA
| | - Christine D. Plant
- Department of Neurosurgery; Stanford Partnership for Spinal Cord Injury and Repair, Stanford University School of Medicine; Stanford California USA
| | - Alan R. Harvey
- School of Anatomy, Physiology and Human Biology; The University of Western Australia; Perth Western Australia Australia
- Western Australian Neuroscience Research Institute; Perth Western Australia Australia
| | - Giles W. Plant
- Department of Neurosurgery; Stanford Partnership for Spinal Cord Injury and Repair, Stanford University School of Medicine; Stanford California USA
| |
Collapse
|
88
|
Trost A, Lange S, Schroedl F, Bruckner D, Motloch KA, Bogner B, Kaser-Eichberger A, Strohmaier C, Runge C, Aigner L, Rivera FJ, Reitsamer HA. Brain and Retinal Pericytes: Origin, Function and Role. Front Cell Neurosci 2016; 10:20. [PMID: 26869887 PMCID: PMC4740376 DOI: 10.3389/fncel.2016.00020] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/18/2016] [Indexed: 12/13/2022] Open
Abstract
Pericytes are specialized mural cells located at the abluminal surface of capillary blood vessels, embedded within the basement membrane. In the vascular network these multifunctional cells fulfil diverse functions, which are indispensable for proper homoeostasis. They serve as microvascular stabilizers, are potential regulators of microvascular blood flow and have a central role in angiogenesis, as they for example regulate endothelial cell proliferation. Furthermore, pericytes, as part of the neurovascular unit, are a major component of the blood-retina/brain barrier. CNS pericytes are a heterogenic cell population derived from mesodermal and neuro-ectodermal germ layers acting as modulators of stromal and niche environmental properties. In addition, they display multipotent differentiation potential making them an intriguing target for regenerative therapies. Pericyte-deficiencies can be cause or consequence of many kinds of diseases. In diabetes, for instance, pericyte-loss is a severe pathological process in diabetic retinopathy (DR) with detrimental consequences for eye sight in millions of patients. In this review, we provide an overview of our current understanding of CNS pericyte origin and function, with a special focus on the retina in the healthy and diseased. Finally, we highlight the role of pericytes in de- and regenerative processes.
Collapse
Affiliation(s)
- Andrea Trost
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and OptometrySalzburg, Austria; Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
| | - Simona Lange
- Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria
| | - Falk Schroedl
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and OptometrySalzburg, Austria; Anatomy, Paracelsus Medical UniversitySalzburg, Austria
| | - Daniela Bruckner
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and Optometry Salzburg, Austria
| | - Karolina A Motloch
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and Optometry Salzburg, Austria
| | - Barbara Bogner
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and Optometry Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and Optometry Salzburg, Austria
| | - Clemens Strohmaier
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and Optometry Salzburg, Austria
| | - Christian Runge
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and Optometry Salzburg, Austria
| | - Ludwig Aigner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria; Anatomy, Paracelsus Medical UniversitySalzburg, Austria
| | - Francisco J Rivera
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria; Anatomy, Paracelsus Medical UniversitySalzburg, Austria
| | - Herbert A Reitsamer
- Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, University Clinic of Ophthalmology and OptometrySalzburg, Austria; Anatomy, Paracelsus Medical UniversitySalzburg, Austria
| |
Collapse
|
89
|
Takayanagi A, Sasaki M, Kataoka-Sasaki Y, Kobayashi K, Matsuda Y, Oka S, Masumori N, Kocsis JD, Honmou O. Intravenous Preload of Mesenchymal Stem Cells Rescues Erectile Function in a Rat Model of Cavernous Nerve Injury. J Sex Med 2015. [PMID: 26211660 DOI: 10.1111/jsm.12957] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
INTRODUCTION We evaluated the potential preventive effects and mechanisms of intravenously preloaded mesenchymal stem cells (MSCs) for erectile dysfunction (ED) in a cavernous nerve (CN) injury model. METHODS Male Sprague-Dawley (SD) rats were used for this study. Rats were randomized into two groups. One group was intravenously preloaded with MSCs (1.0 × 10(6) cells in 1 mL total fluid volume) and the other was infused with medium alone (1 mL Dulbecco's modified Eagle's medium [DMEM]) for sham control, respectively. Crushed CN injury was induced immediately after infusion. The surgeon was blind to the experimental conditions (MSC or medium). MAIN OUTCOME MEASURES To assess erectile function, we measured the intracavernous pressure (ICP) and arterial pressure (AP) at 1 hour and 2 weeks after CN injury. After measuring the initial ICP/AP of pre-injury (normal) male SD rats, they were randomized into the two groups and infused with MSCs or medium. PKH26-labelled MSCs were used for tracking. To investigate the mRNA expression levels of neurotrophins in the major pelvic ganglia (MPG), we performed real-time quantitative real-time polymerase chain reaction. RESULTS The reduction of ICP/AP and area under the curve of ICP (ICP-AUC) in the MSC group was significantly lower than in the DMEM group (P < 0.05; P < 0.05) at 1 hour. The ICP/AP and ICP-AUC at 2 weeks post-injury in the MSC group was significantly higher than in the DMEM group (P < 0.01; P < 0.05). The preloaded PKH26-labelled MSCs were detected in the MPG and CN using confocal microscopy indicating homing of the cells to the injured nerve and ganglia. Glia cell-derived neurotrophic factor (GDNF) and neurturin, which are important neurotrophic factors for erection, had expression levels in MPG significantly higher in the MSC group than in the DMEM group (P < 0.01, 0.05). CONCLUSION Intravenous preload of MSCs before a CN injury may prevent or reduce experimental ED.
Collapse
Affiliation(s)
- Akio Takayanagi
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ko Kobayashi
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yohei Matsuda
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naoya Masumori
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|