1
|
Bjørklund G, Zou L, Peana M, Chasapis CT, Hangan T, Lu J, Maes M. The Role of the Thioredoxin System in Brain Diseases. Antioxidants (Basel) 2022; 11:2161. [PMID: 36358532 PMCID: PMC9686621 DOI: 10.3390/antiox11112161] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 08/08/2023] Open
Abstract
The thioredoxin system, consisting of thioredoxin (Trx), thioredoxin reductase (TrxR), and NADPH, plays a fundamental role in the control of antioxidant defenses, cell proliferation, redox states, and apoptosis. Aberrations in the Trx system may lead to increased oxidative stress toxicity and neurodegenerative processes. This study reviews the role of the Trx system in the pathophysiology and treatment of Alzheimer's, Parkinson's and Huntington's diseases, brain stroke, and multiple sclerosis. Trx system plays an important role in the pathophysiology of those disorders via multiple interactions through oxidative stress, apoptotic, neuro-immune, and pro-survival pathways. Multiple aberrations in Trx and TrxR systems related to other redox systems and their multiple reciprocal relationships with the neurodegenerative, neuro-inflammatory, and neuro-oxidative pathways are here analyzed. Genetic and environmental factors (nutrition, metals, and toxins) may impact the function of the Trx system, thereby contributing to neuropsychiatric disease. Aberrations in the Trx and TrxR systems could be a promising drug target to prevent and treat neurodegenerative, neuro-inflammatory, neuro-oxidative stress processes, and related brain disorders.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610 Mo i Rana, Norway
| | - Lili Zou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania
| | - Jun Lu
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
2
|
Liu C, Hu F, Jiao G, Guo Y, Zhou P, Zhang Y, Zhang Z, Yi J, You Y, Li Z, Wang H, Zhang X. Dental pulp stem cell-derived exosomes suppress M1 macrophage polarization through the ROS-MAPK-NFκB P65 signaling pathway after spinal cord injury. J Nanobiotechnology 2022; 20:65. [PMID: 35109874 PMCID: PMC8811988 DOI: 10.1186/s12951-022-01273-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/17/2022] [Indexed: 12/31/2022] Open
Abstract
Stem cell-derived exosomes have recently been regarded as potential drugs for treating spinal cord injury (SCI) by reducing reactive oxygen species (ROS) and suppressing M1 macrophage polarization. However, the roles of ROS and exosomes in the process of M1 macrophage polarization are not known. Herein, we demonstrated that ROS can induce M1 macrophage polarization and have a concentration-dependent effect. ROS can induce M1 macrophage polarization through the MAPK-NFκB P65 signaling pathway. Dental pulp stem cell (DPSC)-derived exosomes can reduce macrophage M1 polarization through the ROS-MAPK-NFκB P65 signaling pathway in treating SCI. This study suggested that DPSC-derived exosomes might be a potential drug for treating SCI. Disruption of the cycle between ROS and M1 macrophage polarization might also be a potential effective treatment by reducing secondary damage.
Collapse
Affiliation(s)
- Chao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Fanqi Hu
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Genlong Jiao
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Yue Guo
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Pan Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Yuning Zhang
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Zhen Zhang
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Jing Yi
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Yonggang You
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Zhizhong Li
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China.
| | - Hua Wang
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.
| | - Xuesong Zhang
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China.
| |
Collapse
|
3
|
Barzegar M, Stokes KY, Chernyshev O, Kelley RE, Alexander JS. The Role of the ACE2/MasR Axis in Ischemic Stroke: New Insights for Therapy. Biomedicines 2021; 9:1667. [PMID: 34829896 PMCID: PMC8615891 DOI: 10.3390/biomedicines9111667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke remains the leading cause of neurologically based morbidity and mortality. Current stroke treatment is limited to two classes of FDA-approved drugs: thrombolytic agents (tissue plasminogen activator (tPA)) and antithrombotic agents (aspirin and heparin), which have a narrow time-window (<4.5 h) for administration after onset of stroke symptoms. While thrombolytic agents restore perfusion, they carry serious risks for hemorrhage, and do not influence damage responses during reperfusion. Consequently, stroke therapies that can suppress deleterious effects of ischemic injury are desperately needed. Angiotensin converting enzyme-2 (ACE2) has been recently suggested to beneficially influence experimental stroke outcomes by converting the vasoconstrictor Ang II into the vasodilator Ang 1-7. In this review, we extensively discuss the protective functions of ACE2-Ang (1-7)-MasR axis of renin angiotensin system (RAS) in ischemic stroke.
Collapse
Affiliation(s)
- Mansoureh Barzegar
- Molecular and Cellular Physiology, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA; (M.B.); (K.Y.S.)
| | - Karen Y. Stokes
- Molecular and Cellular Physiology, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA; (M.B.); (K.Y.S.)
| | - Oleg Chernyshev
- Neurology, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA; (O.C.); (R.E.K.)
| | - Roger E. Kelley
- Neurology, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA; (O.C.); (R.E.K.)
| | - Jonathan S. Alexander
- Molecular and Cellular Physiology, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA; (M.B.); (K.Y.S.)
- Neurology, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA; (O.C.); (R.E.K.)
- Medicine, LSU Health Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Oral and Maxillofacial Surgery, Ochsner-LSU Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|
4
|
Ogay V, Kumasheva V, Li Y, Mukhlis S, Sekenova A, Olzhayev F, Tsoy A, Umbayev B, Askarova S, Shpekov A, Kaliyev A, Zhetpisbayev B, Makhambetov Y, Akshulakov S, Saparov A, Ramankulov Y. Improvement of Neurological Function in Rats with Ischemic Stroke by Adipose-derived Pericytes. Cell Transplant 2021; 29:963689720956956. [PMID: 32885682 PMCID: PMC7784564 DOI: 10.1177/0963689720956956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pericytes possess high multipotent features and cell plasticity, and produce angiogenic and neurotrophic factors that indicate their high regenerative potential. The aim of this study was to investigate whether transplantation of adipose-derived pericytes can improve functional recovery and neurovascular plasticity after ischemic stroke in rats. Rat adipose-derived pericytes were isolated from subcutaneous adipose tissue by fluorescence-activated cell sorting. Adult male Wistar rats were subjected to 90 min of middle cerebral artery occlusion followed by intravenous injection of rat adipose-derived pericytes 24 h later. Functional recovery evaluations were performed at 1, 7, 14, and 28 days after injection of rat adipose-derived pericytes. Angiogenesis and neurogenesis were examined in rat brains using immunohistochemistry. It was observed that intravenous injection of adipose-derived pericytes significantly improved recovery of neurological function in rats with stroke compared to phosphate-buffered saline-treated controls. Immunohistochemical analysis revealed that the number of blood capillaries was significantly increased along the ischemic boundary zone of the cortex and striatum in stroke rats treated with adipose-derived pericytes. In addition, treatment with adipose-derived pericytes increased the number of doublecortin positive neuroblasts. Our data suggest that transplantation of adipose-derived pericytes can significantly improve the neurologic status and contribute to neurovascular remodeling in rats after ischemic stroke. These data provide a new insight for future cell therapies that aim to treat ischemic stroke patients.
Collapse
Affiliation(s)
- Vyacheslav Ogay
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Venera Kumasheva
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Yelena Li
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Sholpan Mukhlis
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Aliya Sekenova
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan, Kazakhstan
| | - Farkhad Olzhayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Baurzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Azat Shpekov
- Department of Neurosurgery, Medical Centre Hospital of the President's Affairs Administration of the Republic of Kazakhstan, Nur-Sultan, Kazakhstan
| | - Assylbek Kaliyev
- Vascular and Functional Neurosurgery Department, National Center for Neurosurgery, Nur-Sultan, Kazakhstan
| | - Berik Zhetpisbayev
- Vascular and Functional Neurosurgery Department, National Center for Neurosurgery, Nur-Sultan, Kazakhstan
| | - Yerbol Makhambetov
- Vascular and Functional Neurosurgery Department, National Center for Neurosurgery, Nur-Sultan, Kazakhstan
| | - Serik Akshulakov
- Vascular and Functional Neurosurgery Department, National Center for Neurosurgery, Nur-Sultan, Kazakhstan
| | - Arman Saparov
- School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Yerlan Ramankulov
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan, Kazakhstan.,School of Science and Technology, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
5
|
Taurine Augments Telomerase Activity and Promotes Chondrogenesis in Dental Pulp Stem Cells. J Pers Med 2021; 11:jpm11060491. [PMID: 34072707 PMCID: PMC8228366 DOI: 10.3390/jpm11060491] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Stem cell therapy has become an advanced and state-of-the-art procedure to regenerate lost tissues of the human body. Cartilage repair is a challenging task in which stem cells find potential application. One of the important biologic modifiers that can cause chondrogenic differentiation of stem cells is taurine. However, taurine has not been investigated for its effects on dental pulp derived stem cell (DPSC) chondrogenic differentiation. Objective: The objective of the study was to investigate if taurine administration to DPSCs heralds chondrogenic differentiation as ascertained by expression of SOX9, COL2A1, ACAN, ELN, and COMP. The study also investigated if the differentiated cells synthesized glycosaminoglycans, a marker of cartilage formation. The study also aimed to assess proliferative activity of the cells after taurine administration by measuring the hTERT gene and protein expression. Materials and methods: DPSCs were obtained from a molecular biology laboratory and characterization of stem cell markers was done by flow cytometry. The cells were subjected to a MTT assay using various concentrations of taurine. Following this, hTERT gene and protein estimation was done in the control, telomerase inhibitor treated DPSC (TI-III), 10 μM taurine treated DPSC, and TI-III + 10 μM taurine treated DPSCs. A polymerase chain reaction was done to assess gene expression of SOX9, COL2A1, ACAN, ELN, and COMP genes and glycosaminoglycans were estimated in control cells, Induced DPSCs, induced and TI-III treated DPSCs, and 10 μM taurine treated DPSCs. Results: DPSCs expressed CD73, CD90, and CD105 and did not express CD34, CD45, and HLA-DR, which demonstrated that they were mesenchymal stem cells. The MTT assay revealed that various concentrations of taurine did not affect the cell viability of DPSCs. A concentration of 10 μM of taurine was used for further assays. With regard to the hTERT gene and protein expression, the taurine treated cells expressed the highest levels that were statistically significant compared to the other groups. Taurine was also found to restore hTERT expression in telomerase inhibitor treated cells. With regard to chondrogenesis related genes, taurine administration significantly increased the expression of SOX9, COL2A1, ACAN, and ELN genes in DPSCs and caused a significant increase in glycosaminoglycan production by the cells. Conclusions: Taurine can be regarded a biologic modifier that can significantly augment chondrogenic differentiation of DPSCs and can find potential applications in regenerative medicine in the area of cartilage regeneration.
Collapse
|
6
|
von Linstow CU, Hindkjær SM, Nielsen PV, Degn M, Lambertsen KL, Finsen B, Clausen BH. Bone Marrow-Derived IL-1Ra Increases TNF Levels Poststroke. Cells 2021; 10:956. [PMID: 33924148 PMCID: PMC8074385 DOI: 10.3390/cells10040956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor (TNF) and interleukin-1 receptor antagonist (IL-1Ra) are key players in stroke, a disease in which cell-based therapies have shown great potential. Having shown an infarct-reducing effect of bone marrow (BM) cells, especially cells with high IL-1Ra expression, we here investigated the effect of BM cells on TNF and other stroke-related mediators in mice after transient middle cerebral artery occlusion (tMCAo) and in vitro using adult microglial cultures. We analyzed stroke-related genes and inflammatory mediators using qPCR stroke Tier panels, electrochemiluminescence, or enzyme-linked immunosorbent assays. We found a significant correlation and cellular colocalization between microglial-derived TNF and IL-1Ra, though IL-1Ra production was TNF independent. BM treatment significantly increased TNF, interleukin (IL)-10, and IL-4 levels, while C-X-C motif ligand 1 (CXCL1), IL-12p70, and Toll-like receptor 2 (TLR2) decreased, suggesting that BM treatment favors an anti-inflammatory environment. Hierarchical clustering identified Tnf and IL-1rn within the same gene cluster, and subsequent STRING analysis identified TLR2 as a shared receptor. Although IL-1Ra producing BM cells specifically modulated TNF levels, this was TLR2 independent. These results demonstrate BM cells as modulators of poststroke inflammation with beneficial effects on poststroke outcomes and place TNF and IL-1Ra as key players of the defense response after tMCAo.
Collapse
Affiliation(s)
- Christian Ulrich von Linstow
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA;
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.M.H.); (P.V.N.); (K.L.L.); (B.F.)
| | - Sofie Mozart Hindkjær
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.M.H.); (P.V.N.); (K.L.L.); (B.F.)
| | - Pernille Vinther Nielsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.M.H.); (P.V.N.); (K.L.L.); (B.F.)
| | - Matilda Degn
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.M.H.); (P.V.N.); (K.L.L.); (B.F.)
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- BRIDGE—Brain Research—Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.M.H.); (P.V.N.); (K.L.L.); (B.F.)
- BRIDGE—Brain Research—Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.M.H.); (P.V.N.); (K.L.L.); (B.F.)
- BRIDGE—Brain Research—Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
7
|
Liu K, Cai GL, Zhuang Z, Pei SY, Xu SN, Wang YN, Wang H, Wang X, Cui C, Sun MC, Guo SH, Jia KP, Wang XZ, Cai GF. Interleukin-1β-Treated Mesenchymal Stem Cells Inhibit Inflammation in Hippocampal Astrocytes Through Exosome-Activated Nrf-2 Signaling. Int J Nanomedicine 2021; 16:1423-1434. [PMID: 33654394 PMCID: PMC7910114 DOI: 10.2147/ijn.s289914] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interleukin-1β (IL-1)-treated mesenchymal stem cells (MSCs) and IL-1-MSCs-conditioned medium (CM) exert anti-inflammatory roles. Astrocytes are essential for the modulation of synaptic activity and neuronal homeostasis in the brain. Exosomes are the critical mediators in intercellular communication. However, the mechanism underlying the anti-inflammatory effect of IL-1-treated MSCs remains unknown. METHODS In this study, exosomes (IL-1-Exo) were isolated from IL-1-treated MSCs. In addition, lipopolysaccharide (LPS)-treated hippocampal astrocytes and status epilepticus (SE) mice were treated with IL-1-Exo. Inflammatory activity, astrogliosis, and cognitive performance were measured to determine the effect of IL-1-Exo on inflammation. RESULTS The results revealed that IL-1-Exo significantly inhibited LPS-induced astrogliosis and inflammatory responses of astrocytes. Also, IL-1-Exo reversed the LPS-induced effect on calcium signaling. The Nrf2 signaling pathway was associated with the effect of IL-1-Exo in LPS-treated astrocytes. Furthermore, IL-1-Exo reduced the inflammatory response and improved the cognitive performance of SE mice. CONCLUSION The results suggest that IL-1-Exo inhibited LPS-induced inflammatory responses in astrocytes and SE mice and that the effect of IL-1-Exo was primarily mediated through the Nrf-2 signaling pathway. This study provides a new understanding of the molecular mechanism of inflammation-associated brain diseases and an avenue to develop nanotherapeutic agents for the treatment of inflammatory conditions in the brain.
Collapse
Affiliation(s)
- Kai Liu
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Guo-Liang Cai
- Postdoctoral Research Workstation of Harbin Sport University, Harbin, 150001, People’s Republic of China
- Department of Sport Science and Health, Harbin Sport University, Harbin, 150008, People’s Republic of China
| | - Zhe Zhuang
- Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Si-Ying Pei
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Sheng-Nan Xu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Ya-Nan Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Hong Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Xin Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Cheng Cui
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Man-Chao Sun
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Si-Hui Guo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Kun-Ping Jia
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Xiu-Zhen Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Guo-Feng Cai
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| |
Collapse
|
8
|
Li W, Shi L, Hu B, Hong Y, Zhang H, Li X, Zhang Y. Mesenchymal Stem Cell-Based Therapy for Stroke: Current Understanding and Challenges. Front Cell Neurosci 2021; 15:628940. [PMID: 33633544 PMCID: PMC7899984 DOI: 10.3389/fncel.2021.628940] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
Stroke, the most prevalent cerebrovascular disease, causes serious loss of neurological function and is the leading cause of morbidity and mortality worldwide. Despite advances in pharmacological and surgical therapy, treatment for functional rehabilitation following stroke is limited with a consequent serious impact on quality of life. Over the past decades, mesenchymal stem cell (MSCs)-based therapy has emerged as a novel strategy for various diseases including stroke due to their unique properties that include easy isolation, multipotent differentiation potential and strong paracrine capacity. Although MSCs have shown promising results in the treatment of stroke, there remain many challenges to overcome prior to their therapeutic application. In this review, we focus on the following issues: the scientific data from preclinical studies and clinical trials of MSCs in the treatment of stroke; the potential mechanisms underlying MSC-based therapy for stroke; the challenges related to the timing and delivery of MSCs and MSC senescence.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Linli Shi
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Bei Hu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yimei Hong
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Zhang
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuelin Zhang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Li WY, Zhu QB, Jin LY, Yang Y, Xu XY, Hu XY. Exosomes derived from human induced pluripotent stem cell-derived neural progenitor cells protect neuronal function under ischemic conditions. Neural Regen Res 2021; 16:2064-2070. [PMID: 33642395 PMCID: PMC8343330 DOI: 10.4103/1673-5374.308665] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Compared with other stem cells, human induced pluripotent stem cells-derived neural progenitor cells (iPSC-NPCs) are more similar to cortical neurons in morphology and immunohistochemistry. Thus, they have greater potential for promoting the survival and growth of neurons and alleviating the proliferation of astrocytes. Transplantation of stem cell exosomes and stem cells themselves have both been shown to effectively repair nerve injury. However, there is no study on the protective effects of exosomes derived from iPSC-NPCs on oxygen and glucose deprived neurons. In this study, we established an oxygen-glucose deprivation model in embryonic cortical neurons of the rat by culturing the neurons in an atmosphere of 95% N2 and 5% CO2 for 1 hour and then treated them with iPSC-NPC-derived exosomes for 30 minutes. Our results showed that iPSC-NPC-derived exosomes increased the survival of oxygen- and glucose-deprived neurons and the level of brain-derived neurotrophic factor in the culture medium. Additionally, it attenuated oxygen and glucose deprivation-induced changes in the expression of the PTEN/AKT signaling pathway as well as synaptic plasticity-related proteins in the neurons. Further, it increased the length of the longest neurite in the oxygen- and glucose-deprived neurons. These findings validate the hypothesis that exosomes from iPSC-NPCs exhibit a neuroprotective effect on oxygen- and glucose-deprived neurons by regulating the PTEN/AKT signaling pathway and neurite outgrowth. This study was approved by the Animal Ethics Committee of Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China (approval No. SRRSH20191010) on October 10, 2019.
Collapse
Affiliation(s)
- Wen-Yu Li
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Qiong-Bin Zhu
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Lu-Ya Jin
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yi Yang
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xiao-Yan Xu
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Xing-Yue Hu
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine; Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
10
|
Wang L, Deng Z, Yuan R, Zhao Y, Yang M, Hu J, Zhang Y, Li Y, Zhou F, Kang H. Protective effect and mechanism of mesenchymal stem cells on heat stroke induced intestinal injury. Exp Ther Med 2020; 20:3041-3050. [PMID: 32855671 PMCID: PMC7444330 DOI: 10.3892/etm.2020.9051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 05/29/2020] [Indexed: 11/06/2022] Open
Abstract
Heat stroke (HS) is considered to be a severe systemic inflammatory reaction disease that is caused by high fever. The mortality of HS is high worldwide due to the lack of effective treatments. Presently, mesenchymal stem cells (MSCs) have been demonstrated to serve roles in inflammation and immune regulation. Therefore, the current study aimed to investigate the protective effect and mechanism of MSCs against the HS-induced inflammatory response and organ dysfunction. A rat model of HS was induced by a high-temperature environment and treated with MSCs via tail veins. The levels of molecular markers of organ function, inflammatory factors and chemokines were examined at days 1, 7, 14 and 28. Histological staining was performed on the intestines of rats and control groups, and the Chiu's scores of the two groups were compared. The results revealed that MSCs injection significantly reduced the mortality and inhibited the circulatory inflammatory response. Additionally, main organ function, such as in the liver and kidney, were significantly improved following MSCs infusion in HS rats. Furthermore, MSCs treatment significantly improved edema, necrosis and villus exfoliation of intestinal mucosa, and reduced the inflammatory response of intestinal tissue. These results indicated that MSC infusion had therapeutic effects on HS of rats by regulating the circulatory and intestinal inflammatory response. Moreover, MSCs may be able to protect organ function and promote tissue repair in HS. The results of the current study indicated that MSCs may be used as a potential method to treat HS and the resulting organ dysfunction.
Collapse
Affiliation(s)
- Lu Wang
- Medical School of Chinese PLA, Beijing 100853, P.R. China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Zihui Deng
- Department of Biochemistry, Graduate School, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Rui Yuan
- Medical School of Chinese PLA, Beijing 100853, P.R. China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yan Zhao
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Mengmeng Yang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jie Hu
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yu Zhang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yun Li
- Medical School of Chinese PLA, Beijing 100853, P.R. China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Feihu Zhou
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Hongjun Kang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
11
|
Mangin G, Kubis N. Cell Therapy for Ischemic Stroke: How to Turn a Promising Preclinical Research into a Successful Clinical Story. Stem Cell Rev Rep 2020; 15:176-193. [PMID: 30443706 DOI: 10.1007/s12015-018-9864-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stroke is a major public health issue with limited treatment. The pharmacologically or mechanically removing of the clot is accessible to less than 10% of the patients. Stem cell therapy is a promising alternative strategy since it increases the therapeutic time window but many issues remain unsolved. To avoid a new dramatic failure when translating experimental data on the bedside, this review aims to highlight the indispensable checkpoints to make a successful clinical trial based on the current preclinical literature. The large panel of progenitors/ stem cells at the researcher's disposal is to be used wisely, regarding the type of cells, the source of cells, the route of delivery, the time window, since it will directly affect the outcome. Mechanisms are still incompletely understood, although recent studies have focused on the inflammation modulation of most cells types.
Collapse
Affiliation(s)
| | - Nathalie Kubis
- INSERM U965, F-75475, Paris, France. .,Sorbonne Paris Cité, Université Paris Diderot, F-75475, Paris, France. .,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, 2 rue Ambroise Paré, F-75475, Paris, France.
| |
Collapse
|
12
|
Zhang Y, Deng Z, Li Y, Yuan R, Yang M, Zhao Y, Wang L, Zhou F, Kang H. Mesenchymal Stem Cells Provide Neuroprotection by Regulating Heat Stroke-Induced Brain Inflammation. Front Neurol 2020; 11:372. [PMID: 32477247 PMCID: PMC7232542 DOI: 10.3389/fneur.2020.00372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/14/2020] [Indexed: 01/01/2023] Open
Abstract
Heat stroke (HS) is the most acute type of heat illness accompanied with serious central nervous system (CNS) dysfunction. Despite the pathological process being clearly studied, effective treatment is deficient. Currently, mesenchymal stem cells (MSCs) have been demonstrated to have neuroprotective effects as there are no old ones. Thus, the purpose of the present study was to explore the neuroprotective effects and mechanisms of MSCs against HS-induced CNS injury. HS in rat models was induced by a high-temperature environment and treated with MSCs via the tail vein. The results demonstrated that MSC injection significantly reduced the mortality and inhibited the circulation inflammatory response. Moreover, the HS-induced neurological deficit and neuronic damage of the hippocampus were significantly ameliorated by MSC administration. In addition, MSC administration significantly restored astrocytes and inhibited cerebral inflammatory response. These results indicate that MSC infusion has therapeutic effects in HS of rats by regulating the circulation and cerebral inflammatory response. Moreover, astrocytes increased in MSC-treated HS rats when compared with the untreated ones. This may suggest a potential mechanism for HS prevention and therapy through MSC administration.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Zihui Deng
- Biochemistry Department of Graduate School, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Yun Li
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Rui Yuan
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Mengmeng Yang
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Yan Zhao
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Feihu Zhou
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Hongjun Kang
- Department of Critical Care Medicine, General Hospital of the Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
13
|
Shahi M, Abedelahi A, Mohammadnejad D, Rahbarghazi R, Rasta SH, Karimipour M. Exact location of sensorimotor cortex injury after photochemical modulation; evidence of stroke based on stereological and morphometric studies in mice. Lasers Med Sci 2020; 36:91-98. [PMID: 32297252 DOI: 10.1007/s10103-020-03017-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/06/2020] [Indexed: 11/26/2022]
Abstract
The integrity of the structural cerebral cortex is disrupted after stroke either at the macroscopic or microscopic levels. Therefore, many attempts have been gathered to circumvent stroke-associated problems in the brain tissue. The current study was aimed to design an animal model of photochemical stroke using rose bengal (RB) plus laser irradiation (L) after 10, 15, and 20 min (´) and evaluate its effect on the cerebral tissue using unbiased stereological quantitative methods and morphometric histological analysis. Photochemical stroke was induced by intraperitoneal injection of RB dye and further activation through the exposure of the right sensorimotor cortex with the green laser radiation (100 mW; 532 nm). Mice were randomly allocated into 4 groups (each in 15) as follows: control (10 μg/gbw RB), RB + 10'L, RB + 15'L, and RB + 20'L. Target irradiation site was adjusted to 2 mm lateral to the bregma. Vernier caliper morphometric evaluation, cresyl violet staining, and unbiased stereological assays including Cavalier's principle and point counting techniques were used to monitor the pathological changes and lesion volume on days 1, 3, and 7 after the ischemia induction. Our data showed that the mean diameter of the lesion site and lesion infarct volume in the group RB + 20'L) was significantly increased relative to the other groups (P < 0.05). Notably, the lesion volume and diameter in the group RB + 15'L was larger compared with the group RB + 10'L and control mice (P < 0.05). Data showed an increased acute inflammatory response such as hyperemia and edema 3 days after ischemic induction while the intensity of acute changes and lesion volume were reduced and replaced with necrotic and chronic pathological changes including astrogliosis on day 7. It is concluded that the laser irradiation of RB-injected mice at a distinct time period could induce the magnificent degenerative effects on the cerebral cortex which is similar to the stroke condition.
Collapse
Affiliation(s)
- Maryam Shahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, Iran
| | - Daryoush Mohammadnejad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Rasta
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Bioengineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht Street, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Gao J, Bai P, Li Y, Li J, Jia C, Wang T, Zhao H, Si Y, Chen J. Metabolomic Profiling of the Synergistic Effects of Ginsenoside Rg1 in Combination with Neural Stem Cell Transplantation in Ischemic Stroke Rats. J Proteome Res 2020; 19:2676-2688. [PMID: 31968172 DOI: 10.1021/acs.jproteome.9b00639] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jian Gao
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Bai
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuanyuan Li
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingzhong Li
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Caixia Jia
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tieshan Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haibin Zhao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yinchu Si
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
15
|
Hernández R, Jiménez-Luna C, Perales-Adán J, Perazzoli G, Melguizo C, Prados J. Differentiation of Human Mesenchymal Stem Cells towards Neuronal Lineage: Clinical Trials in Nervous System Disorders. Biomol Ther (Seoul) 2020; 28:34-44. [PMID: 31649208 PMCID: PMC6939692 DOI: 10.4062/biomolther.2019.065] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/12/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been proposed as an alternative therapy to be applied into several pathologies of the nervous system. These cells can be obtained from adipose tissue, umbilical cord blood and bone marrow, among other tissues, and have remarkable therapeutic properties. MSCs can be isolated with high yield, which adds to their ability to differentiate into non-mesodermal cell types including neuronal lineage both in vivo and in vitro. They are able to restore damaged neural tissue, thus being suitable for the treatment of neural injuries, and possess immunosuppressive activity, which may be useful for the treatment of neurological disorders of inflammatory etiology. Although the long-term safety of MSC-based therapies remains unclear, a large amount of both pre-clinical and clinical trials have shown functional improvements in animal models of nervous system diseases following transplantation of MSCs. In fact, there are several ongoing clinical trials evaluating the possible benefits this cell-based therapy could provide to patients with neurological damage, as well as their clinical limitations. In this review we focus on the potential of MSCs as a therapeutic tool to treat neurological disorders, summarizing the state of the art of this topic and the most recent clinical studies.
Collapse
Affiliation(s)
- Rosa Hernández
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| | - Cristina Jiménez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges 1066, Switzerland
| | - Jesús Perales-Adán
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain.,Department of Anatomy and Embryology, University of Granada, Granada 18016, Spain.,Biosanitary Institute of Granada (ibs.GRANADA), Granada 18012, Spain
| |
Collapse
|
16
|
Yan J, Goerne T, Zelmer A, Guzman R, Kapfhammer JP, Wellmann S, Zhu X. The RNA-Binding Protein RBM3 Promotes Neural Stem Cell (NSC) Proliferation Under Hypoxia. Front Cell Dev Biol 2019; 7:288. [PMID: 31824945 PMCID: PMC6881237 DOI: 10.3389/fcell.2019.00288] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Neural stem cells (NSCs) reside physiologically in a hypoxic niche to maintain self-renewal and multipotency. Whereas mild hypoxia is known to promote NSC proliferation, severe hypoxia in pathological conditions exerts the reverse effect. The multi-functional RNA-binding protein RBM3 is abundant in NSCs and can be regulated by hypoxic exposure. Although RBM3 has been shown to accelerate cell growth in many cell types, whether and how it affects NSC proliferation in hypoxic environment remains largely unknown. In this study, we tested how RBM3 regulates cell proliferation under hypoxia in C17.2 mouse NSC cell line and in primary mouse NSCs from both the forebrain of postnatal day 0 (P0) mice and the subgranular zone (SGZ) of adult mice. Our results demonstrated that RBM3 expression was highly sensitive to hypoxia, and NSCs were arrested in G0/G1 phase by 5, 2.5, and 1% O2 treatment. When we overexpressed RBM3, hypoxia-induced cell cycle arrest in G0/G1 phase was relieved and more cell transit into S phase was observed. Furthermore, cell viability under hypoxia was also increased by RBM3. In contrast, in RBM3-depleted primary NSCs, less BrdU-incorporated cells were detected, indicating exacerbated cell cycle arrest in G1 to S phase transition. Instead, overexpressed RBM3 significantly increased proliferation ratio in primary NSCs. Our findings indicate RBM3 as a potential target to maintain the proliferation capacity of NSCs under hypoxia, which can be important in NSC-based therapies of acute brain injury and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Jingyi Yan
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Tessa Goerne
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Andrea Zelmer
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Sven Wellmann
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland.,Department of Neonatology, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Xinzhou Zhu
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| |
Collapse
|
17
|
Zhang GL, Zhu ZH, Wang YZ. Neural stem cell transplantation therapy for brain ischemic stroke: Review and perspectives. World J Stem Cells 2019; 11:817-830. [PMID: 31692854 PMCID: PMC6828598 DOI: 10.4252/wjsc.v11.i10.817] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/11/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Brain ischemic stroke is one of the most common causes of death and disability, currently has no efficient therapeutic strategy in clinic. Due to irreversible functional neurons loss and neural tissue injury, stem cell transplantation may be the most promising treatment approach. Neural stem cells (NSCs) as the special type of stem cells only exist in the nervous system, can differentiate into neurons, astrocytes, and oligodendrocytes, and have the abilities to compensate insufficient endogenous nerve cells and improve the inflammatory microenvironment of cell survival. In this review, we focused on the important role of NSCs therapy for brain ischemic stroke, mainly introduced the methods of optimizing the therapeutic efficacy of NSC transplantation, such as transfection and overexpression of specific genes, pretreatment of NSCs with inflammatory factors, and co-transplantation with cytokines. Next, we discussed the potential problems of NSC transplantation which seriously limited their rapid clinical transformation and application. Finally, we expected a new research topic in the field of stem cell research. Based on the bystander effect, exosomes derived from NSCs can overcome many of the risks and difficulties associated with cell therapy. Thus, as natural seed resource of nervous system, NSCs-based cell-free treatment is a newly therapy strategy, will play more important role in treating ischemic stroke in the future.
Collapse
Affiliation(s)
- Gui-Long Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Zhi-Han Zhu
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Ye-Zhong Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| |
Collapse
|
18
|
Airini R, Iordache F, Alexandru D, Savu L, Epureanu FB, Mihailescu D, Amuzescu B, Maniu H. Senescence-induced immunophenotype, gene expression and electrophysiology changes in human amniocytes. J Cell Mol Med 2019; 23:7233-7245. [PMID: 31478614 PMCID: PMC6815807 DOI: 10.1111/jcmm.14495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/23/2022] Open
Abstract
The aim of the study was to evidence replicative senescence‐induced changes in human amniocytes via flow cytometry, quantitative reverse‐transcription‐polymerase chain reaction (qRT‐PCR) and automated/manual patch‐clamp. Both cryopreserved and senescent amniocytes cultured in BIO‐AMF‐2 medium featured high percentages of pluripotency cell surface antigens SSEA‐1, SSEA‐4, TRA1‐60, TRA1‐81 (assessed by flow cytometry) and expression of pluripotency markers Oct4 (Pou5f1) and Nanog (by qRT‐PCR). We demonstrated in senescent vs cryopreserved amniocytes decreases in mesenchymal stem cell surface markers. Senescence‐associated β‐galactosidase stained only senescent amniocytes, and they showed no deoxyuridine incorporation. The gene expression profile revealed a secretory phenotype of senescent amniocytes (increased interleukin (IL)‐1α, IL‐6, IL‐8, transforming growth factor β, nuclear factor κB p65 expression), increases for cell cycle‐regulating genes (p16INK4A), cytoskeletal elements (β‐actin); HMGB1, c‐Myc, Bcl‐2 showed reduced changes and p21, MDM2 decreased. Via patch‐clamp we identified five ion current components: outward rectifier K+ current, an inactivatable component, big conductance Ca2+‐dependent K+ channels (BK) current fluctuations, Na+ current, and inward rectifier K+ current. Iberiotoxin 100 nmol/L blocked 71% of BK fluctuations, and lidocaine 200 μmol/L exerted use‐dependent Na+ current block. Transient receptor potential (TRP)M7‐like current density at −120 mV was significantly increased in senescent amniocytes. The proinflammatory profile acquired by senescent amniocytes in vitro may prevent their use in clinical therapies for immunosuppression, antiapoptotic and healing effects.
Collapse
Affiliation(s)
- Razvan Airini
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Florin Iordache
- Department of Regenerative Medicine, "N. Simionescu" Institute of Cell Biology and Pathology, Bucharest, Romania
| | - Dorin Alexandru
- Department of Regenerative Medicine, "N. Simionescu" Institute of Cell Biology and Pathology, Bucharest, Romania
| | - Lorand Savu
- Genetic Lab S.R.L., Bucharest, Romania.,Fundeni Clinical Institute, Bucharest, Romania
| | - Florin Bogdan Epureanu
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Dan Mihailescu
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Bogdan Amuzescu
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Horia Maniu
- Department of Regenerative Medicine, "N. Simionescu" Institute of Cell Biology and Pathology, Bucharest, Romania
| |
Collapse
|
19
|
Lan X, Sun Z, Chu C, Boltze J, Li S. Dental Pulp Stem Cells: An Attractive Alternative for Cell Therapy in Ischemic Stroke. Front Neurol 2019; 10:824. [PMID: 31428038 PMCID: PMC6689980 DOI: 10.3389/fneur.2019.00824] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is a major cause of disability and mortality worldwide, but effective restorative treatments are very limited at present. Regenerative medicine research revealed that stem cells are promising therapeutic options. Dental pulp stem cells (DPSCs) are autologously applicable cells that origin from the neural crest and exhibit neuro-ectodermal features next to multilineage differentiation potentials. DPSCs are of increasing interest since they are relatively easy to obtain, exhibit a strong proliferation ability, and can be cryopreserved for a long time without losing their multi-directional differentiation capacity. Besides, use of DPSCs can avoid fundamental problems such as immune rejection, ethical controversy, and teratogenicity. Therefore, DPSCs provide a tempting prospect for stroke treatment.
Collapse
Affiliation(s)
- Xiaoyan Lan
- Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, China
| | - Zhengwu Sun
- Department of Pharmacy, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, China
| | - Chengyan Chu
- Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Shen Li
- Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, China
| |
Collapse
|
20
|
Nagpal A, Hillier S, Milton AG, Hamilton-Bruce MA, Koblar SA. PERSPECTIVES: Stroke survivors' views on the design of an early-phase cell therapy trial for patients with chronic ischaemic stroke. Health Expect 2019; 22:1069-1077. [PMID: 31332894 PMCID: PMC6803398 DOI: 10.1111/hex.12932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/19/2019] [Accepted: 05/22/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Stem cell research holds the potential for a paradigm shift in the management of diseases such as stroke. Patient and public involvement in research (PPIR) can bring a focus to issues of clinical relevance and accelerate translation to real-world clinical practice. OBJECTIVE A qualitative thematic analysis of the perspectives of stroke survivors regarding the conduct and design aspects of a proposed phase I clinical cell therapy study in stroke. DESIGN Twelve stroke survivors were purposively recruited in July 2016-August 2017 and participated in semi-structured, face-to-face interviews for input into the design of a proposed phase I clinical study of autologous dental pulp stem cells. Concurrent thematic analysis was conducted until data saturation was achieved. DISCUSSION AND CONCLUSIONS Participants conveyed that the most relevant outcomes to them were regaining participation, decreased dependence on caregivers and improvement in cognition, memory, mood, pain and fatigue. The perception of risk vs. benefit was likely influenced by the time elapsed since stroke, with participants being more willing to accept a higher level of risk early in the post-stroke disease course. They believed that all stroke survivors should be given an opportunity to participate in research, irrespective of their cognitive capacity. A relatively small sample population of 12 stroke survivors was studied as thematic saturation was achieved. PERSPECTIVES study applied principles of PPIR to early-phase cell research. Incorporation of outcomes relevant to patients' need within the study design is critical to generate data that will enable personalized application of regenerative medicine in stroke.
Collapse
Affiliation(s)
- Anjali Nagpal
- Stroke Research Programme, Adelaide Medical School, The University of Adelaide, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Susan Hillier
- Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Austin G Milton
- Stroke Research Programme, Departments of Neurology and Medicine, The Queen Elizabeth Hospital, & Royal Adelaide Hospital, Central Adelaide Local Health Network (CALHN), Adelaide, SA, Australia
| | - Monica A Hamilton-Bruce
- Stroke Research Programme, Adelaide Medical School, The University of Adelaide, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia.,Stroke Research Programme, Departments of Neurology and Medicine, The Queen Elizabeth Hospital, & Royal Adelaide Hospital, Central Adelaide Local Health Network (CALHN), Adelaide, SA, Australia
| | - Simon A Koblar
- Stroke Research Programme, Adelaide Medical School, The University of Adelaide, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, Australia.,Stroke Research Programme, Departments of Neurology and Medicine, The Queen Elizabeth Hospital, & Royal Adelaide Hospital, Central Adelaide Local Health Network (CALHN), Adelaide, SA, Australia
| |
Collapse
|
21
|
Modern Concepts in Regenerative Therapy for Ischemic Stroke: From Stem Cells for Promoting Angiogenesis to 3D-Bioprinted Scaffolds Customized via Carotid Shear Stress Analysis. Int J Mol Sci 2019; 20:ijms20102574. [PMID: 31130624 PMCID: PMC6566983 DOI: 10.3390/ijms20102574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is associated with a tremendous economic and societal burden, and only a few therapies are currently available for the treatment of this devastating disease. The main therapeutic approaches used nowadays for the treatment of ischemic brain injury aim to achieve reperfusion, neuroprotection and neurorecovery. Therapeutic angiogenesis also seems to represent a promising tool to improve the prognosis of cerebral ischemia. This review aims to present the modern concepts and the current status of regenerative therapy for ischemic stroke and discuss the main results of major clinical trials addressing the effectiveness of stem cell therapy for achieving neuroregeneration in ischemic stroke. At the same time, as a glimpse into the future, this article describes modern concepts for stroke prevention, such as the implantation of bioprinted scaffolds seeded with stem cells, whose 3D geometry is customized according to carotid shear stress.
Collapse
|
22
|
Bidirectional Transcriptome Analysis of Rat Bone Marrow-Derived Mesenchymal Stem Cells and Activated Microglia in an In Vitro Coculture System. Stem Cells Int 2018; 2018:6126413. [PMID: 30151012 PMCID: PMC6087576 DOI: 10.1155/2018/6126413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/15/2018] [Accepted: 05/24/2018] [Indexed: 02/07/2023] Open
Abstract
Microglia contribute to the regulation of neuroinflammation and play an important role in the pathogenesis of brain diseases. Thus, regulation of neuroinflammation triggered by activated microglia in brain diseases has become a promising curative strategy. Bone marrow-derived mesenchymal stem cells (BM-MSCs) have been shown to have therapeutic effects, resulting from the regulation of inflammatory conditions in the brain. In this study, we investigated differential gene expression in rat BM-MSCs (rBM-MSCs) that were cocultured with lipopolysaccharide- (LPS-) stimulated primary rat microglia using microarray analysis and evaluated the functional relationships through Ingenuity Pathway Analysis (IPA). We also evaluated the effects of rBM-MSC on LPS-stimulated microglia using a reverse coculture system and the same conditions of the transcriptomic analysis. In the transcriptome of rBM-MSCs, 67 genes were differentially expressed, which were highly related with migration of cells, compared to control. The prediction of the gene network using IPA and experimental validation showed that LPS-stimulated primary rat microglia increase the migration of rBM-MSCs. Reversely, expression patterns of the transcriptome in LPS-stimulated primary rat microglia were changed when cocultured with rBM-MSCs. Our results showed that 65 genes were changed, which were highly related with inflammatory response, compared to absence of rBM-MSCs. In the same way with the aforementioned, the prediction of the gene network and experimental validation showed that rBM-MSCs decrease the inflammatory response of LPS-stimulated primary rat microglia. Our data indicate that LPS-stimulated microglia increase the migration of rBM-MSCs and that rBM-MSCs reduce the inflammatory activity in LPS-stimulated microglia. The results of this study show complex mechanisms underlying the interaction between rBM-MSCs and activated microglia and may be helpful for the development of stem cell-based strategies for brain diseases.
Collapse
|
23
|
Laskowitz DT, Bennett ER, Durham RJ, Volpi JJ, Wiese JR, Frankel M, Shpall E, Wilson JM, Troy J, Kurtzberg J. Allogeneic Umbilical Cord Blood Infusion for Adults with Ischemic Stroke: Clinical Outcomes from a Phase I Safety Study. Stem Cells Transl Med 2018; 7:521-529. [PMID: 29752869 PMCID: PMC6052613 DOI: 10.1002/sctm.18-0008] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/21/2018] [Indexed: 12/16/2022] Open
Abstract
Stroke is a major cause of death and long‐term disability, affecting one in six people worldwide. The only currently available approved pharmacological treatment for ischemic stroke is tissue plasminogen activator; however, relatively few patients are eligible for this therapy. We hypothesized that intravenous (IV) infusion of banked unrelated allogeneic umbilical cord blood (UCB) would improve functional outcomes in patients with ischemic stroke. To investigate this, we conducted a phase I open‐label trial to assess the safety and feasibility of a single IV infusion of non‐human leukocyte antigen (HLA) matched, ABO matched, unrelated allogeneic UCB into adult stroke patients. Ten participants with acute middle cerebral artery ischemic stroke were enrolled. UCB units were matched for blood group antigens and race but not HLA, and infused 3–9 days post‐stroke. The adverse event (AE) profile over a 12 month postinfusion period indicated that the treatment was well‐tolerated in these stroke patients, with no serious AEs directly related to the study product. Study participants were also assessed using neurological and functional evaluations, including the modified Rankin Score (mRS) and National Institute of Health Stroke Scale (NIHSS). At 3 months post‐treatment, all participants had improved by at least one grade in mRS (mean 2.8 ± 0.9) and by at least 4 points in NIHSS (mean 5.9 ± 1.4), relative to baseline. Together, these data suggest that a single i.v. dose of allogeneic non‐HLA matched human UCB cells is safe in adults with ischemic stroke, and support the conduct of a randomized, placebo‐controlled phase 2 study. stemcellstranslationalmedicine2018;7:521–529
Collapse
Affiliation(s)
| | | | - Rebecca J. Durham
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Research Institute/Duke UniversityDurhamNorth CarolinaUSA
| | - John J. Volpi
- Eddy Scurlock Stroke Center, Houston Methodist Neurological InstituteHoustonTexasUSA
| | - Jonathan R. Wiese
- Eddy Scurlock Stroke Center, Houston Methodist Neurological InstituteHoustonTexasUSA
| | - Michael Frankel
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Elizabeth Shpall
- MD Anderson Cancer Center, The University of TexasHoustonTexasUSA
| | - Jeffry M. Wilson
- MD Anderson Cancer Center, The University of TexasHoustonTexasUSA
| | - Jesse Troy
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Research Institute/Duke UniversityDurhamNorth CarolinaUSA
| | - Joanne Kurtzberg
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Research Institute/Duke UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
24
|
Boshuizen MCS, Steinberg GK. Stem Cell-Based Immunomodulation After Stroke: Effects on Brain Repair Processes. Stroke 2018; 49:1563-1570. [PMID: 29724892 DOI: 10.1161/strokeaha.117.020465] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/05/2018] [Accepted: 03/20/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Marieke C S Boshuizen
- From the Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, CA
| | - Gary K Steinberg
- From the Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, CA.
| |
Collapse
|
25
|
Regulated Mesenchymal Stem Cells Mediated Colon Cancer Therapy Assessed by Reporter Gene Based Optical Imaging. Int J Mol Sci 2018; 19:ijms19041002. [PMID: 29584688 PMCID: PMC5979455 DOI: 10.3390/ijms19041002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/24/2018] [Accepted: 03/25/2018] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer is the most common cancer in both men and women and the second most common cause of cancer-related deaths. Suicide gene-based therapy with suicide gene-transduced mesenchymal stem cells (MSCs) is a promising therapeutic strategy. A tetracycline-controlled Tet-On inducible system used to regulate gene expression may be a useful tool for gene-based therapies. The aim of this study was to develop therapeutic MSCs with a suicide gene that is induced by an artificial stimulus, to validate therapeutic gene expression, and to monitor the MSC therapy for colon cancer using optical molecular imaging. For our study, we designed the Tet-On system using a retroviral vector and developed a response plasmid RetroX-TRE (tetracycline response element) expressing a mutant form of herpes simplex virus thymidine kinase (HSV1-sr39TK) with dual reporters (eGFP-Fluc2). Bone marrow-derived MSCs were transduced using a RetroX-Tet3G (Clontech, CA, USA) regulatory plasmid and RetroX-TRE-HSV1-sr39TK-eGFP-IRES-Fluc2, for a system with a Tet-On (MSC-Tet-TK/Fluc2 or MSC-Tet-TK) or without a Tet-On (MSC-TK/Fluc2 or MSC-TK) function. Suicide gene engineered MSCs were co-cultured with colon cancer cells (CT26/Rluc) in the presence of the prodrug ganciclovir (GCV) after stimulation with or without doxycycline (DOX). Treatment efficiency was monitored by assessing Rluc (CT26/Rluc) and Fluc (MSC-Tet-TK and MSC-TK) activity using optical imaging. The bystander effect of therapeutic MSCs was confirmed in CT26/Rluc cells after GCV treatment. Rluc activity in CT26/Rluc cells decreased significantly with GCV treatment of DOX(+) cells (p < 0.05 and 0.01) whereas no significant changes were observed in DOX(-) cells. In addition, Fluc activity in also decreased significantly with DOX(+) MSC-Tet-TK cells, but no signal was observed in DOX(-) cells. In addition, an MSC-TK bystander effect was also confirmed. We assessed therapy with this system in a colon cancer xenograft model (CT26/Rluc). We successfully transduced cells and developed a Tet-On system with the suicide gene HSV1-sr39TK. Our results confirmed the therapeutic efficiency of a suicide gene with the Tet-On system for colon cancer. In addition, our results provide an innovative therapeutic approach using the Tet-On system to eradicate tumors by administration of MSC-Tet-TK cells with DOX and GCV.
Collapse
|
26
|
Redondo-Castro E, Cunningham CJ, Miller J, Brown H, Allan SM, Pinteaux E. Changes in the secretome of tri-dimensional spheroid-cultured human mesenchymal stem cells in vitro by interleukin-1 priming. Stem Cell Res Ther 2018; 9:11. [PMID: 29343288 PMCID: PMC5773162 DOI: 10.1186/s13287-017-0753-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/30/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are one of the most promising candidates for the treatment of major neurological disorders. Desirable therapeutic properties of MSCs include reparative and regenerative potential but, despite their proven safety, the efficacy of MSCs remains controversial. Therefore, it is essential to optimise culture protocols to enhance the therapeutic potential of the MSC secretome. Here we aimed to: assess the increase in secretion of cytokines that may induce repair, regeneration, or immunomodulation when cultured in three dimensions; study the effect of interleukin (IL)-1 priming on two- (2D) and three-dimensional (3D) cultures of MSC; and evaluate the potential use of the modified secretome using microglial-MSC co-cultures. Methods We established a 3D spheroid culture of human MSCs, and compared the secretome in 2D and 3D cultures under primed (IL-1) and unprimed conditions. BV2 microglial cells were stimulated with lipopolysaccharide (LPS) and treated with spheroid conditioned media (CM) or were co-cultured with whole spheroids. Concentrations of secreted cytokines were determined by enzyme-linked immunosorbent assay (ELISA). Protein arrays were used to further evaluate the effect of IL-1 priming in 2D and 3D cultures. Results 3D culture of MSCs significantly increased secretion of the IL-1 receptor antagonist (IL-1Ra), vascular endothelial growth factor (VEGF), and granulocyte-colony stimulating factor (G-CSF) compared with 2D culture, despite priming treatments with IL-1 being more effective in 2D than in 3D. The addition of CM of 3D-MSCs reduced LPS-induced tumour necrosis factor (TNF)-α secretion from BV2 cells, while the 3D spheroid co-cultured with the BV2 cells induced an increase in IL-6, but had no effect on TNF-α release. Protein arrays indicated that priming treatments trigger a more potent immune profile which is necessary to orchestrate an effective tissue repair. This effect was lost in 3D, partly because of the overexpression of IL-6. Conclusions Increased secretion of anti-inflammatory markers occurs when MSCs are cultured in 3D, but this specific secretome did not translate into anti-inflammatory effects on LPS-treated BV2 cells in co-culture. These data highlight the importance of optimising priming treatments and culture conditions to maximise the therapeutic potential of MSC spheroids.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona J Cunningham
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonjo Miller
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Helena Brown
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
27
|
Redondo-Castro E, Cunningham CJ, Miller J, Cain SA, Allan SM, Pinteaux E. Generation of Human Mesenchymal Stem Cell 3D Spheroids Using Low-binding Plates. Bio Protoc 2018; 8:e2968. [PMID: 30294619 DOI: 10.21769/bioprotoc.2968] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The 3D culture of human mesenchymal stem cells (hMSCs) represents a more physiological environment than classical 2D culture and has been used to enhance the MSC secretome or extend cell survival after transplantation. Here we describe a simple and affordable method to generate 3D spheroids of hMSCs by seeding them at high density in a low-binding 96-well plate. Spheroids of hMSCs cultured in low-binding 96-well plates can be used to study the basic biology of the cells and to generate conditioned media or spheroids to be used in transplantation therapeutic approaches. These MSCs or their secretome can be used as a regenerative therapy and for tissue repair across multiple disease areas, including neurodegeneration. In comparison to other methods (hanging drop, use of gels or biomaterials, magnetic levitation, etc.), the method described here is simple and affordable with no need to use specialized equipment, expensive materials or complex reagents.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Division of Neuroscience and Experimental Neurology, Faculty of Biology, Medicine, and Health. University of Manchester, Manchester, UK
| | - Catriona J Cunningham
- Division of Neuroscience and Experimental Neurology, Faculty of Biology, Medicine, and Health. University of Manchester, Manchester, UK
| | - Jonjo Miller
- Division of Neuroscience and Experimental Neurology, Faculty of Biology, Medicine, and Health. University of Manchester, Manchester, UK
| | - Stuart A Cain
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Neurology, Faculty of Biology, Medicine, and Health. University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Division of Neuroscience and Experimental Neurology, Faculty of Biology, Medicine, and Health. University of Manchester, Manchester, UK
| |
Collapse
|
28
|
Zhang T, Yang X, Liu T, Shao J, Fu N, Yan A, Geng K, Xia W. Adjudin-preconditioned neural stem cells enhance neuroprotection after ischemia reperfusion in mice. Stem Cell Res Ther 2017; 8:248. [PMID: 29115993 PMCID: PMC5678778 DOI: 10.1186/s13287-017-0677-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/30/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Transplantation of neural stem cells (NSCs) has been proposed as a promising therapeutic strategy for the treatment of ischemia/reperfusion (I/R)-induced brain injury. However, existing evidence has also challenged this therapy on its limitations, such as the difficulty for stem cells to survive after transplantation due to the unfavorable microenvironment in the ischemic brain. Herein, we have investigated whether preconditioning of NSCs with adjudin, a small molecule compound, could enhance their survivability and further improve the therapeutic effect for NSC-based stroke therapy. METHOD We aimed to examine the effect of adjudin pretreatment on NSCs by measuring a panel of parameters after their transplantation into the infarct area of ipsilateral striatum 24 hours after I/R in mice. RESULTS We found that pretreatment of NSCs with adjudin could enhance the viability of NSCs after their transplantation into the stroke-induced infarct area. Compared with the untreated NSC group, the adjudin-preconditioned group showed decreased infarct volume and neurobehavioral deficiency through ameliorating blood-brain barrier disruption and promoting the expression and secretion of brain-derived neurotrophic factor. We also employed H2O2-induced cell death model in vitro and found that adjudin preconditioning could promote NSC survival through inhibition of oxidative stress and activation of Akt signaling pathway. CONCLUSION This study showed that adjudin could be used to precondition NSCs to enhance their survivability and improve recovery in the stroke model, unveiling the value of adjudin for stem cell-based stroke therapy.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Yang
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Tengyuan Liu
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaxiang Shao
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ningzhen Fu
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Aijuan Yan
- Department of Neurology & Institute of Neurology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Room 211, Med-X Research Institute, 1954 Huashan Road, Shanghai, 200030 China
| | - Keyi Geng
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Department of Neurology & Institute of Neurology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, Room 211, Med-X Research Institute, 1954 Huashan Road, Shanghai, 200030 China
| |
Collapse
|
29
|
Sarmah D, Kaur H, Saraf J, Pravalika K, Goswami A, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Getting Closer to an Effective Intervention of Ischemic Stroke: The Big Promise of Stem Cell. Transl Stroke Res 2017; 9:356-374. [PMID: 29075984 DOI: 10.1007/s12975-017-0580-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
Stem cell therapy for ischemic stroke has widely been explored. Results from both preclinical and clinical studies have immensely supported the judicious use of stem cells as therapy. These provide an attractive means for preserving and replacing the damaged brain tissues following an ischemic attack. Since the past few years, researchers have used various types of stem cells to replenish insulted neuronal and glial cells in neurological disorders. In the present review, we discuss different types of stem cells employed for the treatment of ischemic stroke and mechanisms and challenges these cells face once introduced into the living system. Further, we also present different ways to maneuver and overcome challenges to translate the advances made at the preclinical level to clinics.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Jackson Saraf
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Kanta Pravalika
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Avirag Goswami
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, 382355, India.
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Huang H, Lin F, Jiang J, Chen Y, Mei A, Zhu P. Effects of intra-arterial transplantation of adipose-derived stem cells on the expression of netrin-1 and its receptor DCC in the peri-infarct cortex after experimental stroke. Stem Cell Res Ther 2017; 8:223. [PMID: 29017609 PMCID: PMC5633888 DOI: 10.1186/s13287-017-0671-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 12/16/2022] Open
Abstract
Background Stem cell transplantation has been documented to promote functional recovery in animal models of stroke; however, the underlying mechanisms are not yet fully understood. As netrin-1 and its receptor deleted in colorectal cancer (DCC) are important regulators in neuronal and vascular activities, the present study attempted to explore whether netrin-1 and DCC are involved in the neuroprotection of stem cell-based therapies in a rat ischemic stroke model. Methods Adult male Sprague–Dawley rats were subjected to a transient middle cerebral artery occlusion (MCAO) and subsequently received an intra-arterial injection of 2 × 106 PKH26-labeled adipose-derived stem cells (ADSCs) or saline 24 h later. Neurological function was evaluated by behavioral tests before the rats were sacrificed at days 7 and 14 after MCAO. The migration of ADSCs and regeneration of neuronal fibers and blood vessels were determined by immunofluorescence staining. The expression of netrin-1 and DCC was analyzed by Western blot and immunofluorescence staining. Results ADSC transplantation significantly improved the neurological recovery at days 7 and 14, and noticeably promoted the regeneration of neuronal fibers and blood vessels in the peri-infarct cortex at day 14. PKH26-labeled ADSCs located mainly in the peri-infarct area at days 7 and 14. In ADSC-treated rats, the expression of netrin-1 and DCC significantly increased in the peri-infarct cortex at days 7 and 14. Immunofluorescence staining showed that netrin-1 was mainly expressed by neuronal perikaryal in the peri-infarct cortex, and DCC was mainly expressed by neuronal fibers and was present around the blood vessels in the peri-infarct cortex. Conclusions These findings suggest that ADSC transplantation facilitates the regeneration of neuronal fibers and blood vessels in the peri-infarct cortex and improves neurological functions, which may be attributed, at least in part, to the involvement of upregulated netrin-1 and DCC in the remodeling of neuronal and vascular networks in the peri-infarct cortex.
Collapse
Affiliation(s)
- Huan Huang
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Fan Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Jingjing Jiang
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Yan Chen
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Ainong Mei
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Pengli Zhu
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China. .,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China. .,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
31
|
Redondo-Castro E, Cunningham C, Miller J, Martuscelli L, Aoulad-Ali S, Rothwell NJ, Kielty CM, Allan SM, Pinteaux E. Interleukin-1 primes human mesenchymal stem cells towards an anti-inflammatory and pro-trophic phenotype in vitro. Stem Cell Res Ther 2017; 8:79. [PMID: 28412968 PMCID: PMC5393041 DOI: 10.1186/s13287-017-0531-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/01/2017] [Accepted: 03/08/2017] [Indexed: 12/25/2022] Open
Abstract
Background Inflammation is a key contributor to central nervous system (CNS) injury such as stroke, and is a major target for therapeutic intervention. Effective treatments for CNS injuries are limited and applicable to only a minority of patients. Stem cell-based therapies are increasingly considered for the treatment of CNS disease, because they can be used as in-situ regulators of inflammation, and improve tissue repair and recovery. One promising option is the use of bone marrow-derived mesenchymal stem cells (MSCs), which can secrete anti-inflammatory and trophic factors, can migrate towards inflamed and injured sites or can be implanted locally. Here we tested the hypothesis that pre-treatment with inflammatory cytokines can prime MSCs towards an anti-inflammatory and pro-trophic phenotype in vitro. Methods Human MSCs from three different donors were cultured in vitro and treated with inflammatory mediators as follows: interleukin (IL)-1α, IL-1β, tumour necrosis factor alpha (TNF-α) or interferon-γ. After 24 h of treatment, cell supernatants were analysed by ELISA for expression of granulocyte-colony stimulating factor (G-CSF), IL-10, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), IL-1 receptor antagonist (IL-1Ra) and vascular endothelial growth factor (VEGF). To confirm the anti-inflammatory potential of MSCs, immortalised mouse microglial BV2 cells were treated with bacterial lipopolysaccharide (LPS) and exposed to conditioned media (CM) of naïve or IL-1-primed MSCs, and levels of secreted microglial-derived inflammatory mediators including TNF-α, IL-10, G-CSF and IL-6 were measured by ELISA. Results Unstimulated MSCs constitutively expressed anti-inflammatory cytokines and trophic factors (IL-10, VEGF, BDNF, G-CSF, NGF and IL-1Ra). MSCs primed with IL-1α or IL-1β showed increased secretion of G-CSF, which was blocked by IL-1Ra. Furthermore, LPS-treated BV2 cells secreted less inflammatory and apoptotic markers, and showed increased secretion of the anti-inflammatory IL-10 in response to treatment with CM of IL-1-primed MSCs compared with CM of unprimed MSCs. Conclusions Our results demonstrate that priming MSCs with IL-1 increases expression of trophic factor G-CSF through an IL-1 receptor type 1 (IL-1R1) mechanism, and induces a reduction in the secretion of inflammatory mediators in LPS-activated microglial cells. The results therefore support the potential use of preconditioning treatments of stem cells in future therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0531-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona Cunningham
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonjo Miller
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Licia Martuscelli
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sarah Aoulad-Ali
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Nancy J Rothwell
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cay M Kielty
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
32
|
Delavaran H, Aked J, Sjunnesson H, Lindvall O, Norrving B, Kokaia Z, Lindgren A. Spontaneous Recovery of Upper Extremity Motor Impairment After Ischemic Stroke: Implications for Stem Cell-Based Therapeutic Approaches. Transl Stroke Res 2017; 8:351-361. [PMID: 28205065 PMCID: PMC5493719 DOI: 10.1007/s12975-017-0523-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/29/2017] [Indexed: 01/18/2023]
Abstract
Preclinical studies suggest that stem cell therapy (SCT) may improve sensorimotor recovery after stroke. Upper extremity motor impairment (UEMI) is common after stroke, often entailing substantial disability. To evaluate the feasibility of post-stroke UEMI as a target for SCT, we examined a selected sample of stroke patients potentially suitable for SCT, aiming to assess the frequency and recovery of UEMI, as well as its relation to activity limitations and participation restrictions. Patients aged 20–75 years with first-ever ischemic stroke, and National Institutes of Health Stroke Scale (NIHSS) scores 1–18, underwent brain diffusion-weighted MRI within 4 days of stroke onset (n = 108). Survivors were followed up after 3–5 years, including assessment with NIHSS, Fugl-Meyer assessment of upper extremity (FMA-UE), modified Rankin Scale (mRS), and Stroke Impact Scale (SIS). UEMI was defined as NIHSS arm/hand score ≥1. UEMI recovery was evaluated with change in NIHSS arm/hand scores between baseline and follow-up. Of 97 survivors, 84 were available to follow-up. Among 76 subjects (of 84) without recurrent stroke, 41 had UEMI at baseline of which 10 had residual UEMI at follow-up. The FMA-UE showed moderate-severe impairment in seven of 10 survivors with residual UEMI. UEMI was correlated to mRS (rs = 0.49, p < 0.001) and the SIS social participation domain (rs = −0.38, p = 0.001). Nearly 25% of the subjects with UEMI at baseline had residual impairment after 3–5 years, whereas about 75% showed complete recovery. Most of the subjects with residual UEMI had moderate-severe impairment, which correlated strongly to dependency in daily activities and social participation restrictions. Our findings suggest that SCT targeting post-stroke UEMI may be clinically valuable with significant meaningful benefits for patients but also emphasize the need of early prognostication to detect patients that will have residual impairment in order to optimize patient selection for SCT.
Collapse
Affiliation(s)
- Hossein Delavaran
- Department of Clinical Sciences Lund, Division of Neurology, Lund University, Lund, Sweden.
- Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Lund, Sweden.
| | - Joseph Aked
- Department of Clinical Sciences Lund, Division of Neurology, Lund University, Lund, Sweden
| | - Håkan Sjunnesson
- Center for Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Olle Lindvall
- Department of Clinical Sciences Lund, Division of Neurology, Lund University, Lund, Sweden
- Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Lund, Sweden
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Bo Norrving
- Department of Clinical Sciences Lund, Division of Neurology, Lund University, Lund, Sweden
- Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Lund, Sweden
| | - Zaal Kokaia
- Department of Clinical Sciences Lund, Division of Neurology, Lund University, Lund, Sweden
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Arne Lindgren
- Department of Clinical Sciences Lund, Division of Neurology, Lund University, Lund, Sweden
- Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
33
|
Zhang L, Tao W, Feng H, Chen Y. Transcriptional and Genomic Targets of Neural Stem Cells for Functional Recovery after Hemorrhagic Stroke. Stem Cells Int 2017; 2017:2412890. [PMID: 28133486 PMCID: PMC5241497 DOI: 10.1155/2017/2412890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/21/2016] [Indexed: 01/27/2023] Open
Abstract
Hemorrhagic stroke is a life-threatening disease characterized by a sudden rupture of cerebral blood vessels, and it is widely believed that neural cell death occurs after exposure to blood metabolites or subsequently damaged cells. Neural stem cells (NSCs), which maintain neurogenesis and are found in subgranular zone and subventricular zone, are thought to be an endogenous neuroprotective mechanism for these brain injuries. However, due to the complexity of NSCs and their microenvironment, current strategies cannot satisfactorily enhance functional recovery after hemorrhagic stroke. It is well known that transcriptional and genomic pathways play important roles in ensuring the normal functions of NSCs, including proliferation, migration, differentiation, and neural reconnection. Recently, emerging evidence from the use of new technologies such as next-generation sequencing and transcriptome profiling has provided insight into our understanding of genomic function and regulation of NSCs. In the present article, we summarize and present the current data on the control of NSCs at both the transcriptional and genomic levels. Using bioinformatics methods, we sought to predict novel therapeutic targets of endogenous neurogenesis and exogenous NSC transplantation for functional recovery after hemorrhagic stroke, which could also advance our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Le Zhang
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Wenjing Tao
- College of Computer and Information Science, Southwest University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
34
|
Choi TM, Yun M, Lee JK, Park JT, Park MS, Kim HS. Proteomic Analysis of a Rat Cerebral Ischemic Injury Model after Human Cerebral Endothelial Cell Transplantation. J Korean Neurosurg Soc 2016; 59:544-550. [PMID: 27847565 PMCID: PMC5106351 DOI: 10.3340/jkns.2016.59.6.544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 02/08/2023] Open
Abstract
Objective Cerebral endothelial cells have unique biological features and are fascinating candidate cells for stroke therapy. Methods In order to understand the molecular mechanisms of human cerebral endothelial cell (hCMEC/D3) transplantation in a rat stroke model, we performed proteomic analysis using 2-dimensional electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Protein expression was confirmed by quantitative real-time PCR and Western blot. Results Several protein spots were identified by gel electrophoresis in the sham, cerebral ischemia (CI), and CI with hCMEC/D3 treatment cerebral ischemia with cell transplantation (CT) groups, and we identified 14 differentially expressed proteins in the CT group. Proteins involved in mitochondrial dysfunction (paraplegin matrix AAA peptidase subunit, SPG7), neuroinflammation (peroxiredoxin 6, PRDX6), and neuronal death (zinc finger protein 90, ZFP90) were markedly reduced in the CT group compared with the CI group. The expression of chloride intracellular channel 4 proteins involved in post-ischemic vasculogenesis was significantly decreased in the CI group but comparable to sham in the CT group. Conclusion These results contribute to our understanding of the early phase processes that follow cerebral endothelial cell treatment in CI. Moreover, some of the identified proteins may present promising new targets for stroke therapy.
Collapse
Affiliation(s)
- Tae-Min Choi
- Department of Neurosurgery, Gwangju Christian Hospital, Gwangju, Korea.; Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Misun Yun
- Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Jung-Kil Lee
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Jong-Tae Park
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Man-Seok Park
- Department of Neurosurgery, Chonnam National University Medical School, Gwangju, Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Korea.; Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
35
|
Hsuan YCY, Lin CH, Chang CP, Lin MT. Mesenchymal stem cell-based treatments for stroke, neural trauma, and heat stroke. Brain Behav 2016; 6:e00526. [PMID: 27781140 PMCID: PMC5064338 DOI: 10.1002/brb3.526] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/05/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation has been reported to improve neurological function following neural injury. Many physiological and molecular mechanisms involving MSC therapy-related neuroprotection have been identified. METHODS A review is presented of articles that pertain to MSC therapy and diverse brain injuries including stroke, neural trauma, and heat stroke, which were identified using an electronic search (e.g., PubMed), emphasize mechanisms of MSC therapy-related neuroprotection. We aim to discuss neuroprotective mechanisms that underlie the beneficial effects of MSCs in treating stroke, neural trauma, and heatstroke. RESULTS MSC therapy is promising as a means of augmenting brain repair. Cell incorporation into the injured tissue is not a prerequisite for the beneficial effects exerted by MSCs. Paracrine signaling is believed to be the most important mediator of MSC therapy in brain injury. The multiple mechanisms of action of MSCs include enhanced angiogenesis and neurogenesis, immunomodulation, and anti-inflammatory effects. Microglia are the first source of the inflammatory cascade during brain injury. Cytokines, including tumor necrosis factor-α, interleukin-1β, and interleukin-6, are significantly produced by microglia in the brain after experimental brain injury. The proinflammatory M1 phenotype of microglia is associated with tissue destruction, whereas the anti-inflammatory M2 phenotype of microglia facilitates repair and regeneration. MSC therapy may improve outcomes of ischemic stroke, neural trauma, and heatstroke by inhibiting the activity of M1 phenotype of microglia but augmenting the activity of M2 phenotype of microglia. CONCLUSION This review offers a testable platform for targeting microglial-mediated cytokines in clinical trials based upon the rational design of MSC therapy in the future. MSCs that are derived from the placenta provide a great choice for stem cell therapy. Although targeting the microglial activation is an important approach to reduce the burden of the injury, it is not the only one. This review focuses on this specific aspect.
Collapse
Affiliation(s)
| | | | - Ching-Ping Chang
- Department of Medical Research Chi Mei Medical Center Tainan Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research Chi Mei Medical Center Tainan Taiwan
| |
Collapse
|
36
|
Gaining Mechanistic Insights into Cell Therapy Using Magnetic Resonance Imaging. CURRENT STEM CELL REPORTS 2016. [DOI: 10.1007/s40778-016-0059-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
37
|
In Vivo Targeted MR Imaging of Endogenous Neural Stem Cells in Ischemic Stroke. Molecules 2016; 21:molecules21091143. [PMID: 27589699 PMCID: PMC6273863 DOI: 10.3390/molecules21091143] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 08/19/2016] [Accepted: 08/26/2016] [Indexed: 12/22/2022] Open
Abstract
Acute ischemic stroke remains a leading cause of death and disability. Endogenous neurogenesis enhanced via activation of neural stem cells (NSCs) could be a promising method for stroke treatment. In vivo targeted tracking is highly desirable for monitoring the dynamics of endogenous NSCs in stroke. Previously, we have successfully realized in vivo targeted MR imaging of endogenous NSCs in normal adult mice brains by using anti-CD15 antibody-conjugated superparamagnetic iron oxide nanoparticles (anti-CD15-SPIONs) as the molecular probe. Herein, we explore the performance of this molecular probe in targeted in vivo tracking of activated endogenous NSCs in ischemic stroke. Our study showed that intraventricular injection of anti-CD15-SPIONs could label activated endogenous NSCs in situ seven days after ischemic stroke, which were detected as enlarged areas of hypo-intense signals on MR imaging at 7.0 T. The treatment of cytosine arabinosine could inhibit the activation of endogenous NSCs, which was featured by the disappearance of areas of hypo-intense signals on MR imaging. Using anti-CD15-SPIONs as imaging probes, the dynamic process of activation of endogenous NSCs could be readily monitored by in vivo MR imaging. This targeted imaging strategy would be of great benefit to develop a new therapeutic strategy utilizing endogenous NSCs for ischemic stroke.
Collapse
|
38
|
Winderlich JN, Kremer KL, Koblar SA. Adult human dental pulp stem cells promote blood-brain barrier permeability through vascular endothelial growth factor-a expression. J Cereb Blood Flow Metab 2016; 36:1087-97. [PMID: 26661186 PMCID: PMC4908623 DOI: 10.1177/0271678x15608392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/30/2015] [Indexed: 01/09/2023]
Abstract
Stem cell therapy is a promising new treatment option for stroke. Intravascular administration of stem cells is a valid approach as stem cells have been shown to transmigrate the blood-brain barrier. The mechanism that causes this effect has not yet been elucidated. We hypothesized that stem cells would mediate localized discontinuities in the blood-brain barrier, which would allow passage into the brain parenchyma. Here, we demonstrate that adult human dental pulp stem cells express a soluble factor that increases permeability across an in vitro model of the blood-brain barrier. This effect was shown to be the result of vascular endothelial growth factor-a. The effect could be amplified by exposing dental pulp stem cell to stromal-derived factor 1, which stimulates vascular endothelial growth factor-a expression. These findings support the use of dental pulp stem cell in therapy for stroke.
Collapse
Affiliation(s)
- Joshua N Winderlich
- Stroke Research Programme, School of Medicine, University of Adelaide, Adelaide, Australia Centre for Stem Cell Research, Robinson Institute, Adelaide, Australia
| | - Karlea L Kremer
- Stroke Research Programme, School of Medicine, University of Adelaide, Adelaide, Australia Centre for Stem Cell Research, Robinson Institute, Adelaide, Australia
| | - Simon A Koblar
- Stroke Research Programme, School of Medicine, University of Adelaide, Adelaide, Australia Centre for Stem Cell Research, Robinson Institute, Adelaide, Australia Department of Neurology, Queen Elizabeth Hospital, Woodville, Australia
| |
Collapse
|
39
|
Clausen BH, Lambertsen KL, Dagnæs-Hansen F, Babcock AA, von Linstow CU, Meldgaard M, Kristensen BW, Deierborg T, Finsen B. Cell therapy centered on IL-1Ra is neuroprotective in experimental stroke. Acta Neuropathol 2016; 131:775-91. [PMID: 26860727 PMCID: PMC4835531 DOI: 10.1007/s00401-016-1541-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/28/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022]
Abstract
Cell-based therapies are emerging as new promising treatments in stroke. However, their functional mechanism and therapeutic potential during early infarct maturation has so far received little attention. Here, we asked if cell-based delivery of the interleukin-1 receptor antagonist (IL-1Ra), a known neuroprotectant in stroke, can promote neuroprotection, by modulating the detrimental inflammatory response in the tissue at risk. We show by the use of IL-1Ra-overexpressing and IL-1Ra-deficient mice that IL-1Ra is neuroprotective in stroke. Characterization of the cellular and spatiotemporal production of IL-1Ra and IL-1α/β identifies microglia, not infiltrating leukocytes, as the major sources of IL-1Ra after experimental stroke, and shows IL-1Ra and IL-1β to be produced by segregated subsets of microglia with a small proportion of these cells co-expressing IL-1α. Reconstitution of whole body irradiated mice with IL-1Ra-producing bone marrow cells is associated with neuroprotection and recruitment of IL-1Ra-producing leukocytes after stroke. Neuroprotection is also achieved by therapeutic injection of IL-1Ra-producing bone marrow cells 30 min after stroke onset, additionally improving the functional outcome in two different stroke models. The IL-1Ra-producing bone marrow cells increase the number of IL-1Ra-producing microglia, reduce the availability of IL-1β, and modulate mitogen-activated protein kinase (MAPK) signaling in the ischemic cortex. The importance of these results is underlined by demonstration of IL-1Ra-producing cells in the human cortex early after ischemic stroke. Taken together, our results attribute distinct neuroprotective or neurotoxic functions to segregated subsets of microglia and suggest that treatment strategies increasing the production of IL-1Ra by infiltrating leukocytes or microglia may also be neuroprotective if applied early after stroke onset in patients.
Collapse
|
40
|
Doeppner TR, Traut V, Heidenreich A, Kaltwasser B, Bosche B, Bähr M, Hermann DM. Conditioned Medium Derived from Neural Progenitor Cells Induces Long-term Post-ischemic Neuroprotection, Sustained Neurological Recovery, Neurogenesis, and Angiogenesis. Mol Neurobiol 2016; 54:1531-1540. [PMID: 26860410 DOI: 10.1007/s12035-016-9748-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/26/2016] [Indexed: 12/15/2022]
Abstract
Adult neural progenitor cells (NPCs) induce post-ischemic long-term neuroprotection and brain remodeling by releasing of survival- and plasticity-promoting mediators. To evaluate whether secreted factors may mimic neuroprotective and restorative effects of NPCs, we exposed male C57BL6 mice to focal cerebral ischemia and intravenously applied conditioned medium (CM) derived from subventricular zone NPCs. CM dose-dependently reduced infarct volume and brain leukocyte infiltration after 48 h when delivered up to 12 h after focal cerebral ischemia. Neuroprotection persisted in the post-acute stroke phase yielding enhanced neurological recovery that lasted throughout the 28-day observation period. Increased Bcl-2, phosphorylated Akt and phosphorylated STAT-3 abundance, and reduced caspase-3 activity and Bax abundance were noted in ischemic brains of CM-treated mice at 48 h post-stroke, indicative of enhanced cell survival signaling. Long-term neuroprotection was associated with increased brain glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF) concentrations at 28 days resulting in increased neurogenesis and angiogenesis. The observation that NPC-derived CM induces sustained neuroprotection and neurological recovery suggests that cell transplantation may be dispensable when secreted factors are instead administered.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany. .,Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey. .,Department of Neurology, University of Göttingen Medical School, Göttingen, Germany.
| | - Viktorija Traut
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Alexander Heidenreich
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Britta Kaltwasser
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Bert Bosche
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany.,Department of Surgery, Division of Neurosurgery, St Michael's Hospital, Keenan Research Centre for Biomedical Science and the Li Ka Shing Knowledge Institute of St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Mathias Bähr
- Department of Neurology, University of Göttingen Medical School, Göttingen, Germany
| | - Dirk M Hermann
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany
| |
Collapse
|
41
|
Madhu V, Dighe AS, Cui Q, Deal DN. Dual Inhibition of Activin/Nodal/TGF-β and BMP Signaling Pathways by SB431542 and Dorsomorphin Induces Neuronal Differentiation of Human Adipose Derived Stem Cells. Stem Cells Int 2015; 2016:1035374. [PMID: 26798350 PMCID: PMC4699250 DOI: 10.1155/2016/1035374] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/03/2015] [Indexed: 12/16/2022] Open
Abstract
Damage to the nervous system can cause devastating diseases or musculoskeletal dysfunctions and transplantation of progenitor stem cells can be an excellent treatment option in this regard. Preclinical studies demonstrate that untreated stem cells, unlike stem cells activated to differentiate into neuronal lineage, do not survive in the neuronal tissues. Conventional methods of inducing neuronal differentiation of stem cells are complex and expensive. We therefore sought to determine if a simple, one-step, and cost effective method, previously reported to induce neuronal differentiation of embryonic stem cells and induced-pluripotent stem cells, can be applied to adult stem cells. Indeed, dual inhibition of activin/nodal/TGF-β and BMP pathways using SB431542 and dorsomorphin, respectively, induced neuronal differentiation of human adipose derived stem cells (hADSCs) as evidenced by formation of neurite extensions, protein expression of neuron-specific gamma enolase, and mRNA expression of neuron-specific transcription factors Sox1 and Pax6 and matured neuronal marker NF200. This process correlated with enhanced phosphorylation of p38, Erk1/2, PI3K, and Akt1/3. Additionally, in vitro subcutaneous implants of SB431542 and dorsomorphin treated hADSCs displayed significantly higher expression of active-axonal-growth-specific marker GAP43. Our data offers novel insights into cell-based therapies for the nervous system repair.
Collapse
Affiliation(s)
- Vedavathi Madhu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Abhijit S. Dighe
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Quanjun Cui
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - D. Nicole Deal
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
42
|
Modern stem cell therapy: approach to disease. Wien Klin Wochenschr 2015; 127 Suppl 5:S199-203. [PMID: 26659705 DOI: 10.1007/s00508-015-0903-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
Abstract
Various types of stem cells exist, each with their own advantages and disadvantages. Considering the current available evidence, important preclinical and clinical studies regarding the therapeutic potential of stem cells, stem cell therapy might be the important strategy for tissue repair. The development of stem cell therapy for tissue repair has primarily relied on stem cells, especially mesenchymal stem cells. Multilineage differentiation into all of the described cells are considered as important candidates for a range of diseases like neurological diseases, cardiovascular diseases, gastrointestinal cancer and genetic defects, as well as for acute and chronic wounds healing and pharmaceutical treatment. We review the properties and multipotency of stem cells and their differentiation potential, once cultured under specific growth conditions, for use in cell-based therapies and functional tissue replacement.
Collapse
|
43
|
Marlier Q, Verteneuil S, Vandenbosch R, Malgrange B. Mechanisms and Functional Significance of Stroke-Induced Neurogenesis. Front Neurosci 2015; 9:458. [PMID: 26696816 PMCID: PMC4672088 DOI: 10.3389/fnins.2015.00458] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/16/2015] [Indexed: 01/01/2023] Open
Abstract
Stroke affects one in every six people worldwide, and is the leading cause of adult disability. After stroke, some limited spontaneous recovery occurs, the mechanisms of which remain largely unknown. Multiple, parallel approaches are being investigated to develop neuroprotective, reparative and regenerative strategies for the treatment of stroke. For years, clinical studies have tried to use exogenous cell therapy as a means of brain repair, with varying success. Since the rediscovery of adult neurogenesis and the identification of adult neural stem cells in the late nineties, one promising field of investigation is focused upon triggering and stimulating this self-repair system to replace the neurons lost following brain injury. For instance, it is has been demonstrated that the adult brain has the capacity to produce large numbers of new neurons in response to stroke. The purpose of this review is to provide an updated overview of stroke-induced adult neurogenesis, from a cellular and molecular perspective, to its impact on brain repair and functional recovery.
Collapse
Affiliation(s)
- Quentin Marlier
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman Liege, Belgium
| | | | - Renaud Vandenbosch
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman Liege, Belgium
| | - Brigitte Malgrange
- GIGA-Neurosciences, University of Liege, C.H.U. Sart Tilman Liege, Belgium
| |
Collapse
|
44
|
Stevanato L, Hicks C, Sinden JD. Differentiation of a Human Neural Stem Cell Line on Three Dimensional Cultures, Analysis of MicroRNA and Putative Target Genes. J Vis Exp 2015:52410. [PMID: 25938519 PMCID: PMC4541566 DOI: 10.3791/52410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells (NSCs) are capable of self-renewal and differentiation into neurons, astrocytes and oligodendrocytes under specific local microenvironments. In here, we present a set of methods used for three dimensional (3D) differentiation and miRNA analysis of a clonal human neural stem cell (hNSC) line, currently in clinical trials for stroke disability (NCT01151124 and NCT02117635, Clinicaltrials.gov). HNSCs were derived from an ethical approved first trimester human fetal cortex and conditionally immortalized using retroviral integration of a single copy of the c-mycER(TAM)construct. We describe how to measure axon process outgrowth of hNSCs differentiated on 3D scaffolds and how to quantify associated changes in miRNA expression using PCR array. Furthermore we exemplify computational analysis with the aim of selecting miRNA putative targets. SOX5 and NR4A3 were identified as suitable miRNA putative target of selected significantly down-regulated miRNAs in differentiated hNSC. MiRNA target validation was performed on SOX5 and NR4A3 3'UTRs by dual reporter plasmid transfection and dual luciferase assay.
Collapse
|
45
|
Nucci LP, Silva HR, Giampaoli V, Mamani JB, Nucci MP, Gamarra LF. Stem cells labeled with superparamagnetic iron oxide nanoparticles in a preclinical model of cerebral ischemia: a systematic review with meta-analysis. Stem Cell Res Ther 2015; 6:27. [PMID: 25889904 PMCID: PMC4425914 DOI: 10.1186/s13287-015-0015-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 11/26/2014] [Accepted: 02/23/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Although there is an increase in clinical trials assessing the efficacy of cell therapy in structural and functional regeneration after stroke, there are not enough data in the literature describing the best cell type to be used, the best route, and also the best nanoparticle to analyze these stem cells in vivo. This review analyzed published data on superparamagnetic iron oxide nanoparticle (SPION)-labeled stem cells used for ischemic stroke therapy. Method We performed a systematic review and meta-analysis of data from experiments testing the efficacy of cellular treatment with SPION versus no treatment to improve behavioral or modified neural scale outcomes in animal models of stroke by the Cochrane Collaboration and indexed in EMBASE, PubMed, and Web of Science since 2000. To test the impact of study quality and design characteristics, we used random-effects meta-regression. In addition, trim and fill were used to assess publication bias. Results The search retrieved 258 articles. After application of the inclusion criteria, 24 reports published between January 2000 and October 2014 were selected. These 24 articles were analyzed for nanoparticle characteristics, stem cell types, and efficacy in animal models. Conclusion This study highlights the therapeutic role of stem cells in stroke and emphasizes nanotechnology as an important tool for monitoring stem cell migration to the affected neurological locus.
Collapse
Affiliation(s)
- Leopoldo P Nucci
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil. .,Universidade Federal de São Paulo, Rua Sena Madureira, 1500 - Vila Clementino, 04021-001, São Paulo-SP, Brazil.
| | - Helio R Silva
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil. .,Santa Casa Misericórdia de São Paulo, Dr. Cesario Motta Junior, 61 - Vila Buarque, 01221-020, São Paulo-SP, Brazil.
| | - Viviana Giampaoli
- Instituto de Matemática e Estatística, Universidade de São Paulo, Rua do Matão 1010 - Cidade Universitária, 05508-090, São Paulo-SP, Brazil.
| | - Javier B Mamani
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil.
| | - Mariana P Nucci
- LIM44, Universidade de São Paulo, Rua Dr Éneas de Carvalho Aguiar, 255 - Cerqueira César, 05403-000, São Paulo-SP, Brazil.
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil. .,Universidade Federal de São Paulo, Rua Sena Madureira, 1500 - Vila Clementino, 04021-001, São Paulo-SP, Brazil. .,Santa Casa Misericórdia de São Paulo, Dr. Cesario Motta Junior, 61 - Vila Buarque, 01221-020, São Paulo-SP, Brazil.
| |
Collapse
|
46
|
Ikegame Y, Yamashita K, Nakashima S, Nomura Y, Yonezawa S, Asano Y, Shinoda J, Hara H, Iwama T. Fate of graft cells: what should be clarified for development of mesenchymal stem cell therapy for ischemic stroke? Front Cell Neurosci 2014; 8:322. [PMID: 25374506 PMCID: PMC4204523 DOI: 10.3389/fncel.2014.00322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/24/2014] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are believed to be promising for cell administration therapy after ischemic stroke. Because of their advantageous characteristics, such as ability of differentiation into neurovascular lineages, avoidance of immunological problems, and abundance of graft cells in mesodermal tissues, studies regarding MSC therapy have increased recently. However, several controversies are yet to be resolved before a worldwide consensus regarding a standard protocol is obtained. In particular, the neuroprotective effects, the rate of cell migration to the lesion, and differentiation direction differ depending on preclinical observations. Analyses of these differences and application of recent developments in stem cell biology or engineering in imaging modality may contribute to identification of criteria for optimal stem cell therapy in which reliable protocols, which control cell quality and include safe administration procedures, are defined for each recovery phase after cerebral ischemia. In this mini review, we examine controversies regarding the fate of grafts and the prospects for advanced therapy that could be obtained through recent developments in stem cell research as direct conversion to neural cells.
Collapse
Affiliation(s)
- Yuka Ikegame
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan ; Department of Cell Signaling, Gifu University Graduate School of Medicine Gifu, Japan
| | - Kentaro Yamashita
- Department of Neurosurgery, Gifu University Graduate School of Medicine Gifu, Japan ; Department of Neurosurgery, Murakami Memorial Hospital, Asahi University Gifu, Japan
| | - Shigeru Nakashima
- Department of Cell Signaling, Gifu University Graduate School of Medicine Gifu, Japan
| | - Yuichi Nomura
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan
| | - Shingo Yonezawa
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan
| | - Yoshitaka Asano
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan
| | - Jun Shinoda
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan
| | - Hideaki Hara
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine Gifu, Japan
| |
Collapse
|
47
|
Shichita T, Ito M, Yoshimura A. Post-ischemic inflammation regulates neural damage and protection. Front Cell Neurosci 2014; 8:319. [PMID: 25352781 PMCID: PMC4196547 DOI: 10.3389/fncel.2014.00319] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/23/2014] [Indexed: 12/31/2022] Open
Abstract
Post-ischemic inflammation is important in ischemic stroke pathology. However, details of the inflammation process, its resolution after stroke and its effect on pathology and neural damage have not been clarified. Brain swelling, which is often fatal in ischemic stroke patients, occurs at an early stage of stroke due to endothelial cell injury and severe inflammation by infiltrated mononuclear cells including macrophages, neutrophils, and lymphocytes. At early stage of inflammation, macrophages are activated by molecules released from necrotic cells [danger-associated molecular patterns (DAMPs)], and inflammatory cytokines and mediators that increase ischemic brain damage by disruption of the blood–brain barrier are released. After post-ischemic inflammation, macrophages function as scavengers of necrotic cell and brain tissue debris. Such macrophages are also involved in tissue repair and neural cell regeneration by producing tropic factors. The mechanisms of inflammation resolution and conversion of inflammation to neuroprotection are largely unknown. In this review, we summarize information accumulated recently about DAMP-induced inflammation and the neuroprotective effects of inflammatory cells, and discuss next generation strategies to treat ischemic stroke.
Collapse
Affiliation(s)
- Takashi Shichita
- Department of Microbiology and Immunology, School of Medicine, Keio University Tokyo, Japan ; Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency Tokyo, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, School of Medicine, Keio University Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, School of Medicine, Keio University Tokyo, Japan
| |
Collapse
|
48
|
Effects of acute versus post-acute systemic delivery of neural progenitor cells on neurological recovery and brain remodeling after focal cerebral ischemia in mice. Cell Death Dis 2014; 5:e1386. [PMID: 25144721 PMCID: PMC4454329 DOI: 10.1038/cddis.2014.359] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 12/17/2022]
Abstract
Intravenous transplantation of neural progenitor cells (NPCs) induces functional recovery after stroke, albeit grafted cells are not integrated into residing neural networks. However, a systematic analysis of intravenous NPC delivery at acute and post-acute time points and their long-term consequences does not exist. Male C57BL6 mice were exposed to cerebral ischemia, and NPCs were intravenously grafted on day 0, on day 1 or on day 28. Animals were allowed to survive for up to 84 days. Mice and tissues were used for immunohistochemical analysis, flow cytometry, ELISA and behavioral tests. Density of grafted NPCs within the ischemic hemisphere was increased when cells were transplanted on day 28 as compared with transplantation on days 0 or 1. Likewise, transplantation on day 28 yielded enhanced neuronal differentiation rates of grafted cells. Post-ischemic brain injury, however, was only reduced when NPCs were grafted at acute time points. On the contrary, reduced post-ischemic functional deficits due to NPC delivery were independent of transplantation paradigms. NPC-induced neuroprotection after acute cell delivery was due to stabilization of the blood–brain barrier (BBB), reduction in microglial activation and modulation of both peripheral and central immune responses. On the other hand, post-acute NPC transplantation stimulated post-ischemic regeneration via enhanced angioneurogenesis and increased axonal plasticity. Acute NPC delivery yields long-term neuroprotection via enhanced BBB integrity and modulation of post-ischemic immune responses, whereas post-acute NPC delivery increases post-ischemic angioneurogenesis and axonal plasticity. Post-ischemic functional recovery, however, is independent of NPC delivery timing, which offers a broad therapeutic time window for stroke treatment.
Collapse
|