1
|
Schallmoser A, Einenkel R, Färber C, Hüren V, Emrich N, John J, Sänger N. Comparison of angiogenic potential in vitrified vs. slow frozen human ovarian tissue. Sci Rep 2023; 13:12885. [PMID: 37558708 PMCID: PMC10412559 DOI: 10.1038/s41598-023-39920-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023] Open
Abstract
Vitrification of ovarian tissue is a promising alternative approach to the traditional slow freezing method. Few empirical investigations have been conducted to determine the angiogenic profiles of these two freezing methods. In this study we aimed to answer the question whether one of the cryopreservation methods should be preferred based on the secretion of angiogenic factors. Tissue culture with reduced oxygen (5%) was conducted for 48 h with samples of fresh, slow frozen/thawed and vitrified/rapid warmed ovarian cortex tissue from 20 patients. From each patient, tissue was used in all three treatment groups. Tissue culture supernatants were determined regarding cytokine expression profiles of angiogenin, angiopoietin-2, epidermal growth factor, basic fibroblast growth factor, heparin binding epidermal growth factor, hepatocyte growth factor, Leptin, Platelet-derived growth factor B, placental growth factor and vascular endothelial growth factor A via fluoroimmunoassay. Apoptotic changes were assessed by TUNEL staining of cryosections and supplemented by hematoxylin and eosin and proliferating cell nuclear antigen staining. Comparing the angiogenic expression profiles of vitrified/rapid warmed tissue with slow frozen/thawed tissue samples, no significant differences were observed. Detection of apoptotic DNA fragmentation via TUNEL indicated minor apoptotic profiles that were not significantly different comparing both cryopreservation methods. Vitrification of ovarian cortical tissue does not appear to impact negatively on the expression profile of angiogenic factors and may be regarded as an effective alternative approach to the traditional slow freezing method.
Collapse
Affiliation(s)
- Andreas Schallmoser
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| | - Rebekka Einenkel
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Cara Färber
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Vanessa Hüren
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Norah Emrich
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Julia John
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Nicole Sänger
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
2
|
Bhuiyan MH, Clarkson AN, Ali MA. Optimization of thermoresponsive chitosan/β-glycerophosphate hydrogels for injectable neural tissue engineering application. Colloids Surf B Biointerfaces 2023; 224:113193. [PMID: 36773410 DOI: 10.1016/j.colsurfb.2023.113193] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Regeneration of neural tissue and recovery of lost functions following an accident or disease to the central nervous system remains a major challenge worldwide, with limited treatment options available. The main reason for the failure of conventional therapeutic techniques to regenerate neural tissue is the presence of blood-brain barrier separating nervous system from systemic circulation and the limited capacity of self-regeneration of the nervous system. Injectable hydrogels have shown great promise for neural tissue engineering given their suitability for minimally invasive in situ delivery and tunable mechanical and biological properties. Chitosan (CS)/β-glycerophosphate (β-GP) hydrogels have been extensively investigated and shown regenerative potential in a wide variety of tissues such as bone and cartilage tissue engineering. However, the potential of CS/β-GP hydrogels has never been tested for injectable neural tissue engineering applications. In the present study, CS/β-GP hydrogels, consisting of 0.5-2% CS and 2-3% β-GP, were prepared and characterized to investigate their suitability for injectable neural tissue engineering applications. The resulting CS/β-GP-hydrogels showed a varying range of properties depending on the CS/β-GP blend ratio. In particular, the 0.5%:3% and 0.75%:3% CS/β-GP hydrogels underwent rapid gelation (3 min and 5 min, respectively) at physiological temperature (37 °C) and pH (7.4). They also had suitable porosity, osmolality, swelling behavior and biodegradation for tissue engineering. The biocompatibility of hydrogels was determined in vitro using PC12 cells, an immortalized cell line with neuronal cell-like properties, revealing that these hydrogels supported cell growth and proliferation. In conclusion, the thermoresponsive 0.5%:3% and 0.75%:3% CS/β-GP hydrogels had the greatest potential for neural tissue engineering.
Collapse
Affiliation(s)
- Mozammel Haque Bhuiyan
- Center for Bioengineering and Nanomedicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Sir John Walsh Research Institute, Faculty of Dentistry, Division of Health Science, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - M Azam Ali
- Center for Bioengineering and Nanomedicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Sir John Walsh Research Institute, Faculty of Dentistry, Division of Health Science, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
3
|
Ho E, Deng Y, Akbar D, Da K, Létourneau M, Morshead CM, Chatenet D, Shoichet MS. Tunable Surface Charge Enables the Electrostatic Adsorption-Controlled Release of Neuroprotective Peptides from a Hydrogel-Nanoparticle Drug Delivery System. ACS APPLIED MATERIALS & INTERFACES 2023; 15:91-105. [PMID: 36520607 DOI: 10.1021/acsami.2c17631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
We exploit the electrostatic interactions between the positively charged neuroprotective peptide, pituitary adenylate cyclase-activating polypeptide (PACAP), and negatively charged poly(lactic-co-glycolic acid) (PLGA) nanoparticles to control PACAP release from the surface of nanoparticles dispersed in a hyaluronan-methylcellulose (HAMC) hydrogel composite. PACAP is a promising therapeutic for the treatment of neurological disorders, yet it has been difficult to deliver in vivo. Herein, the PACAP release rate was tuned by manipulating peptide adsorption onto the surface of blank nanoparticles by modifying either nanoparticle loading in the hydrogel or nanoparticle surface charge. This peptide-nanoparticle interaction was controlled by the pH-responsive carboxylic acid end terminal groups of PLGA. We further validated this system with the controlled release of a novel stabilized PACAP analog: Ac-[Ala15, Ala20]PACAP-propylamide, which masks its recognition to peptidases in circulation. Both wild-type and stabilized PACAP released from the vehicle increased the production of neuroprotective Interleukin-6 from cultured primary astrocytes. Using computational fluid dynamics methods, PACAP release from the composite was predicted based on experimentally derived adsorption isotherms, which exhibited similar release profiles to experimental data. This versatile adsorption-based system was used to deliver PACAP locally to the brains of stroke-injured mice over a 10 day period in vivo, highlighting its effectiveness for the controlled release of PACAP to the central nervous system.
Collapse
Affiliation(s)
- Eric Ho
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, OntarioM5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Yaoqi Deng
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Dania Akbar
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Kevin Da
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Myriam Létourneau
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QuebecH7 V 1B7, Canada
| | - Cindi M Morshead
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, OntarioM5S 3G9, Canada
- Department of Surgery, University of Toronto, 149 College Street, Toronto, OntarioM5S 3E1, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, OntarioM5S 3E1, Canada
| | - David Chatenet
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QuebecH7 V 1B7, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, OntarioM5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, OntarioM5S 3E1, Canada
| |
Collapse
|
4
|
Zhang SY, Zhao J, Ni JJ, Li H, Quan ZZ, Qing H. Application and prospects of high-throughput screening for in vitro neurogenesis. World J Stem Cells 2022; 14:393-419. [PMID: 35949394 PMCID: PMC9244953 DOI: 10.4252/wjsc.v14.i6.393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, high-throughput screening (HTS) has made great contributions to new drug discovery. HTS technology is equipped with higher throughput, minimized platforms, more automated and computerized operating systems, more efficient and sensitive detection devices, and rapid data processing systems. At the same time, in vitro neurogenesis is gradually becoming important in establishing models to investigate the mechanisms of neural disease or developmental processes. However, challenges remain in generating more mature and functional neurons with specific subtypes and in establishing robust and standardized three-dimensional (3D) in vitro models with neural cells cultured in 3D matrices or organoids representing specific brain regions. Here, we review the applications of HTS technologies on in vitro neurogenesis, especially aiming at identifying the essential genes, chemical small molecules and adaptive microenvironments that hold great prospects for generating functional neurons or more reproductive and homogeneous 3D organoids. We also discuss the developmental tendency of HTS technology, e.g., so-called next-generation screening, which utilizes 3D organoid-based screening combined with microfluidic devices to narrow the gap between in vitro models and in vivo situations both physiologically and pathologically.
Collapse
Affiliation(s)
- Shu-Yuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Jun-Jun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
5
|
Thomas JM, Louca I, Bolan F, Sava O, Allan SM, Lawrence CB, Pinteaux E. Regenerative Potential of Hydrogels for Intracerebral Hemorrhage: Lessons from Ischemic Stroke and Traumatic Brain Injury Research. Adv Healthc Mater 2021; 10:e2100455. [PMID: 34197036 PMCID: PMC11468990 DOI: 10.1002/adhm.202100455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/15/2021] [Indexed: 01/02/2023]
Abstract
Intracerebral hemorrhage (ICH) is a deadly and debilitating type of stroke, caused by the rupture of cerebral blood vessels. To date, there are no restorative interventions approved for use in ICH patients, highlighting a critical unmet need. ICH shares some pathological features with other acute brain injuries such as ischemic stroke (IS) and traumatic brain injury (TBI), including the loss of brain tissue, disruption of the blood-brain barrier, and activation of a potent inflammatory response. New biomaterials such as hydrogels have been recently investigated for their therapeutic benefit in both experimental IS and TBI, owing to their provision of architectural support for damaged brain tissue and ability to deliver cellular and molecular therapies. Conversely, research on the use of hydrogels for ICH therapy is still in its infancy, with very few published reports investigating their therapeutic potential. Here, the published use of hydrogels in experimental ICH is commented upon and how approaches reported in the IS and TBI fields may be applied to ICH research to inform the design of future therapies is described. Unique aspects of ICH that are distinct from IS and TBI that should be considered when translating biomaterial-based therapies between disease models are also highlighted.
Collapse
Affiliation(s)
- Josephine M. Thomas
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Irene Louca
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Faye Bolan
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Oana‐Roxana Sava
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Catherine B. Lawrence
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| | - Emmanuel Pinteaux
- Geoffrey Jefferson Brain Research CentreThe Manchester Academic Health Science CentreNorthern Care Alliance NHS GroupThe University of ManchesterManchesterM13 9PTUK
- Division of Neuroscience and Experimental PsychologyFaculty of BiologyMedicine and HealthThe University of ManchesterManchesterM13 9PTUK
| |
Collapse
|
6
|
Ucar B. Natural biomaterials in brain repair: A focus on collagen. Neurochem Int 2021; 146:105033. [PMID: 33785419 DOI: 10.1016/j.neuint.2021.105033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials derived from natural resources have increasingly been used for versatile applications in the central nervous system (CNS). Thanks to their biocompatibility and biodegradability, natural biomaterials offer vast possibilities for future clinical repair strategies for the CNS. These materials can be used for diverse applications such as hydrogels to fill the tissue cavities, microparticles to deliver drugs across the blood-brain barrier, and scaffolds to transplant stem cells. In this review, various uses of prominent protein and polysaccharide biomaterials, with a special focus on collagen, in repair and regenerative applications for the brain are summarized together with their individual advantages and disadvantages.
Collapse
Affiliation(s)
- Buket Ucar
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
7
|
Ali MA, Bhuiyan MH. Types of biomaterials useful in brain repair. Neurochem Int 2021; 146:105034. [PMID: 33789130 DOI: 10.1016/j.neuint.2021.105034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/28/2021] [Accepted: 03/22/2021] [Indexed: 01/21/2023]
Abstract
Biomaterials is an emerging field in the study of brain tissue engineering and repair or neurogenesis. The fabrication of biomaterials that can replicate the mechanical and viscoelastic features required by the brain, including the poroviscoelastic responses, force dissipation, and solute diffusivity are essential to be mapped from the macro to the nanoscale level under physiological conditions in order for us to gain an effective treatment for neurodegenerative diseases. This research topic has identified a critical study gap that must be addressed, and that is to source suitable biomaterials and/or create reliable brain-tissue-like biomaterials. This chapter will define and discuss the various types of biomaterials, their structures, and their function-properties features which would enable the development of next-generation biomaterials useful in brain repair.
Collapse
Affiliation(s)
- M Azam Ali
- Center for Bioengineering and Nanomedicine, Faculty of Dentistry, Division of Health Sciences, University of Otago, Dunedin, New Zealand.
| | - Mozammel Haque Bhuiyan
- Center for Bioengineering and Nanomedicine, Faculty of Dentistry, Division of Health Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
8
|
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: 4.8] [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
|
9
|
Kandalam S, De Berdt P, Ucakar B, Vanvarenberg K, Bouzin C, Gratpain V, Diogenes A, Montero-Menei CN, des Rieux A. Human dental stem cells of the apical papilla associated to BDNF-loaded pharmacologically active microcarriers (PAMs) enhance locomotor function after spinal cord injury. Int J Pharm 2020; 587:119685. [PMID: 32712253 DOI: 10.1016/j.ijpharm.2020.119685] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023]
Abstract
There is no treatment for spinal cord injury (SCI) that fully repairs the damages. One strategy is to inject mesenchymal stem cells around the lesion to benefit from their immunomodulatory properties and neuroprotective effect. Our hypothesis was that the combination of dental stem cells from the apical papilla (SCAP) with pharmacologically active microcarriers (PAMs) releasing brain-derived neurotrophic factor (BDNF) would improve rat locomotor function by immunomodulation and neuroprotection. BDNF-PAMs were prepared by solid/oil/water emulsion of poly(L-lactide-co-glycolide) and nanoprecipitated BDNF and subsequent coating with fibronectin. SCAP were then seeded on BDNF-PAMs. SCAP expression of neuronal and immunomodulatory factors was evaluated in vitro. SCAP BDNF-PAMs were injected in a rat spinal cord contusion model and their locomotor function was evaluated by Basso, Beattie, and Bresnahan (BBB) scoring. Impact on inflammation and neuroprotection/axonal growth was evaluated by immunofluorescence. Culture on PAMs induced the overexpression of immunomodulatory molecules and neural/neuronal markers. Injection of SCAP BDNF-PAMs at the lesion site improved rat BBB scoring, reduced the expression of inducible nitric oxide synthase and increased the expression of βIII tubulin, GAP43, and 5-HT. These results confirm the suitability and versatility of PAMs as combined drug and cell delivery system for regenerative medicine applications but also that BDNF-PAMs potentialize the very promising therapeutic potential of SCAP in the scope of SCI.
Collapse
Affiliation(s)
- Saikrishna Kandalam
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Bruxelles, Belgium; CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers F-49933, France
| | - Pauline De Berdt
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Bruxelles, Belgium
| | - Bernard Ucakar
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Bruxelles, Belgium
| | - Kevin Vanvarenberg
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Bruxelles, Belgium
| | - Caroline Bouzin
- IREC Imaging platform (2IP), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, UCLouvain, IREC, 1200 Brussels, Belgium
| | - Viridiane Gratpain
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Bruxelles, Belgium
| | - Anibal Diogenes
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | - Anne des Rieux
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Bruxelles, Belgium.
| |
Collapse
|
10
|
Rai N, Singh AK, Singh SK, Gaurishankar B, Kamble SC, Mishra P, Kotiya D, Barik S, Atri N, Gautam V. Recent technological advancements in stem cell research for targeted therapeutics. Drug Deliv Transl Res 2020; 10:1147-1169. [DOI: 10.1007/s13346-020-00766-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
11
|
Bai WF, Zhang Y, Xu W, Li W, Li M, Yuan F, Luo X, Zhang M. Isolation and Characterization of Neural Progenitor Cells From Bone Marrow in Cell Replacement Therapy of Brain Injury. Front Cell Neurosci 2020; 14:49. [PMID: 32226361 PMCID: PMC7080866 DOI: 10.3389/fncel.2020.00049] [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: 12/18/2019] [Accepted: 02/21/2020] [Indexed: 12/27/2022] Open
Abstract
Many studies supported that bone marrow mesenchymal stem cells (BM-MSCs) can differentiate into neural cells, but few researchers detected mature and function of nerve cells, especially in vivo study. Some researchers even suggested that BM-MSCs transplantation would not be able to differentiate into functional neural cells. To figure out the dispute, this study examined bone marrow-derived sphere-like cells, harvested via neural stem cell suspension culture, then identified as bone marrow-derived neural progenitor cells (BM-NPCs) by finding the expression of neural progenitor cells genes and proteins, neural progenitor cells characteristic and nerve cell differentiation induced through both methods. Moreover, BM-NPCs transplantation showed long-term survival and improved the ethological and histological indexes of brain injury rats, demonstrating functional nervous cells differentiated from BM-NPCs. These in vitro and in vivo results confirmed BM-NPCs differentiating into mature and functional nerve cells. This study provided valuable experimental data for BM-NPCs, suggesting a potential alternative treatment of central nervous injury disease.
Collapse
Affiliation(s)
- Wen-fang Bai
- Department of Rehabilitation Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, China
| | - Yuling Zhang
- School of Medical Instrument and Food Engineering, The University of Shanghai for Science and Technology, Shanghai, China
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, a Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
| | - Weicheng Xu
- Department of Rehabilitation Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, China
| | - Weikun Li
- Department of Rehabilitation Medicine, Zengcheng District People’s Hospital of Guangzhou, Guangzhou, China
| | - Meihui Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Fengying Yuan
- Department of Rehabilitation Medicine, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, China
- Shenzhen Sanming Project Group, Spaulding Rehabilitation Hospital, a Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
| | - Mingsheng Zhang
- Department of Rehabilitation Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, China
- *Correspondence: Mingsheng Zhang
| |
Collapse
|
12
|
Bolan F, Louca I, Heal C, Cunningham CJ. The Potential of Biomaterial-Based Approaches as Therapies for Ischemic Stroke: A Systematic Review and Meta-Analysis of Pre-clinical Studies. Front Neurol 2019; 10:924. [PMID: 31507524 PMCID: PMC6718570 DOI: 10.3389/fneur.2019.00924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/09/2019] [Indexed: 01/07/2023] Open
Abstract
Background: In recent years pre-clinical stroke research has shown increased interest in the development of biomaterial-based therapies to promote tissue repair and functional recovery. Such strategies utilize biomaterials as structural support for tissue regeneration or as delivery vehicles for therapeutic agents. While a range of biomaterials have been tested in stroke models, currently no overview is available for evaluating the benefit of these approaches. We therefore performed a systematic review and meta-analysis of studies investigating the use of biomaterials for the treatment of stroke in experimental animal models. Methods: Studies were identified by searching electronic databases (PubMed, Web of Science) and reference lists of relevant review articles. Studies reporting lesion volume and/or neurological score were included. Standardized mean difference (SMD) and 95% confidence intervals were calculated using DerSimonian and Laird random effects. Study quality and risk of bias was assessed using the CAMARADES checklist. Publication bias was visualized by funnel plots followed by trim and fill analysis of missing publications. Results: A total of 66 publications were included in the systematic review, of which 44 (86 comparisons) were assessed in the meta-analysis. Overall, biomaterial-based interventions improved both lesion volume (SMD: -2.98, 95% CI: -3.48, -2.48) and neurological score (SMD: -2.3, 95% CI: -2.85, -1.76). The median score on the CAMARADES checklist was 5.5/10 (IQR 4.25-6). Funnel plots of lesion volume and neurological score data revealed pronounced asymmetry and publication bias. Additionally, trim and fill analysis estimated 19 "missing" studies for the lesion volume outcome adjusting the effect size to -1.91 (95% CI: -2.44, -1.38). Conclusions: Biomaterials including scaffolds and particles exerted a positive effect on histological and neurological outcomes in pre-clinical stroke models. However, heterogeneity in the field, publication bias and study quality scores which may be another source of bias call for standardization of outcome measures and improved study reporting.
Collapse
Affiliation(s)
- Faye Bolan
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Irene Louca
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Calvin Heal
- Faculty of Biology, Medicine and Health, Centre for Biostatistics, Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Catriona J. Cunningham
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom,*Correspondence: Catriona J. Cunningham
| |
Collapse
|
13
|
Influencing neuroplasticity in stroke treatment with advanced biomaterials-based approaches. Adv Drug Deliv Rev 2019; 148:204-218. [PMID: 30579882 DOI: 10.1016/j.addr.2018.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/05/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023]
Abstract
Since the early 1990s, we have known that the adult brain is not static and has the capacity to repair itself. The delivery of various therapeutic factors and cells have resulted in some exciting pre-clinical and clinical outcomes in stroke models by targeting post-injury plasticity to enhance recovery. Developing a deeper understanding of the pathways that modulate plasticity will enable us to optimize delivery strategies for therapeutics and achieve more robust effects. Biomaterials are a key tool for the optimization of these potential treatments, owing to their biocompatibility and tunability. In this review, we identify factors and targets that impact plastic processes known to contribute to recovery, discuss the role of biomaterials in enhancing the efficacy of treatment strategies, and suggest combinatorial approaches based on the stage of injury progression.
Collapse
|
14
|
Tang JD, Lampe KJ. From de novo peptides to native proteins: advancements in biomaterial scaffolds for acute ischemic stroke repair. Biomed Mater 2018; 13:034103. [DOI: 10.1088/1748-605x/aaa4c3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
bFGF plays a neuroprotective role by suppressing excessive autophagy and apoptosis after transient global cerebral ischemia in rats. Cell Death Dis 2018; 9:172. [PMID: 29416039 PMCID: PMC5833346 DOI: 10.1038/s41419-017-0229-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/20/2017] [Accepted: 12/14/2017] [Indexed: 12/31/2022]
Abstract
Transient global cerebral ischemia (tGCI) is a cerebrovascular disorder that can cause apoptotic neuronal damage and functional deficits. Basic fibroblast growth factor (bFGF) was reported to be highly expressed in the central nervous system (CNS) and to exert neuroprotective effects against different CNS diseases. However, the effects of bFGF on tGCI have not been studied intensively. This study was conducted to investigate the effect of bFGF and its underlying mechanism in an animal model of tGCI. After intracerebroventricular (i.c.v.) injection of bFGF, functional improvement was observed, and the number of viable neurons increased in the ischemia-vulnerable hippocampal CA1 region. Apoptosis was induced after tGCI and could be attenuated by bFGF treatment via inhibition of p53 mitochondrial translocation. In addition, autophagy was activated during this process, and bFGF could inhibit activation of autophagy through the mTOR pathway. Rapamycin, an activator of autophagy, was utilized to explore the relationship among bFGF, apoptosis, and autophagy. Apoptosis deteriorated after rapamycin treatment, which indicated that excessive autophagy could contribute to the apoptosis process. In conclusion, these results demonstrate that bFGF could exert neuroprotective effects in the hippocampal CA1 region by suppressing excessive autophagy via the mTOR pathway and inhibiting apoptosis by preventing p53 mitochondrial translocation. Furthermore, our results suggest that bFGF may be a promising therapeutic agent to for treating tGCI in response to major adverse events, including cardiac arrest, shock, extracorporeal circulation, traumatic hemorrhage, and asphyxiation.
Collapse
|
16
|
Huang B, Jiang XC, Zhang TY, Hu YL, Tabata Y, Chen Z, Pluchino S, Gao JQ. Peptide modified mesenchymal stem cells as targeting delivery system transfected with miR-133b for the treatment of cerebral ischemia. Int J Pharm 2017; 531:90-100. [PMID: 28827201 DOI: 10.1016/j.ijpharm.2017.08.073] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/12/2017] [Accepted: 08/12/2017] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have been regarded as potential targeting vehicles and demonstrated to exert therapeutic benefits for brain diseases. Direct homing to diseased tissue is crucial for stem cell-based therapy. In this study, a peptide-based targeting approach was established to enhance cell homing to cerebral ischemic lesion. Palmitic acid-peptide painted onto the cell membrane was able to direct MSCs to ischemic tissues without any observed cell cytotoxicity and influence on differentiation, thus reducing accumulation of cells in peripheral organs and increasing engraftment of cells in the targeted tissues. With enhanced cell homing, MSCs were used to deliver miR-133b to increase the expression level of miR-133b in an ischemic lesion and further improve therapeutic effects. This study is the first to develop MSCs co-modified with targeting peptide and microRNAs as potential targeting therapeutic agents. This targeting delivery system is expected to be applicable to other cell types and other diseases aside from stroke.
Collapse
Affiliation(s)
- Bing Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Xin-Chi Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Tian-Yuan Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yu-Lan Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridgeshire, UK
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
17
|
Fakoya AOJ. New Delivery Systems of Stem Cells for Vascular Regeneration in Ischemia. Front Cardiovasc Med 2017; 4:7. [PMID: 28286751 PMCID: PMC5323391 DOI: 10.3389/fcvm.2017.00007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 02/07/2017] [Indexed: 01/08/2023] Open
Abstract
The finances of patients and countries are increasingly overwhelmed with the plague of cardiovascular diseases as a result of having to chronically manage the associated complications of ischemia such as heart failures, neurological deficits, chronic limb ulcers, gangrenes, and amputations. Hence, scientific research has sought for alternate therapies since pharmacological and surgical treatments have fallen below expectations in providing the desired quality of life. The advent of stem cells research has raised expectations with respect to vascular regeneration and tissue remodeling, hence assuring the patients of the possibility of an improved quality of life. However, these supposed encouraging results have been short-lived as the retention, survival, and engraftment rates of these cells appear to be inadequate; hence, the long-term beneficial effects of these cells cannot be ascertained. These drawbacks have led to the relentless research into better ways to deliver stem cells or angiogenic factors (which mobilize stem cells) to the regions of interest to facilitate increased retention, survival, engraftment, and regeneration. This review considered methods, such as the use of scaffolds, retrograde coronary delivery, improved combinations, stem cell pretreatment, preconditioning, stem cell exosomes, mannitol, magnet, and ultrasound-enhanced delivery, homing techniques, and stem cell modulation. Furthermore, the study appraised the possibility of a combination therapy of stem cells and macrophages, considering the enormous role macrophages play in repair, remodeling, and angiogenesis.
Collapse
|
18
|
He Y, Jin X, Wang J, Meng M, Hou Z, Tian W, Li Y, Wang W, Wei Y, Wang Y, Meng H, Lu X, Chen Z, Fu L. Umbilical cord-derived mesenchymal stem cell transplantation for treating elderly vascular dementia. Cell Tissue Bank 2017; 18:53-59. [DOI: 10.1007/s10561-017-9609-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 01/13/2017] [Indexed: 02/06/2023]
|
19
|
Modification of Bone Marrow Stem Cells for Homing and Survival During Cerebral Ischemia. BONE MARROW STEM CELL THERAPY FOR STROKE 2017. [PMCID: PMC7121342 DOI: 10.1007/978-981-10-2929-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Over the last decade, major advances have been made in stem cell-based therapy for ischemic stroke, which is one of the leading causes of death and disability worldwide. Various stem cells from bone marrow, such as mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), and endothelial progenitor cells (EPCs), have shown therapeutic potential for stroke. Concomitant with these exciting findings are some fundamental bottlenecks that must be overcome in order to accelerate their clinical translation, including the low survival and engraftment caused by the harsh microenvironment after transplantation. In this chapter, strategies such as gene modification, hypoxia/growth factor preconditioning, and biomaterial-based methods to improve cell survival and homing are summarized, and the potential strategies for their future application are also discussed.
Collapse
|
20
|
Spiller KL, Vunjak-Novakovic G. Clinical translation of controlled protein delivery systems for tissue engineering. Drug Deliv Transl Res 2016; 5:101-15. [PMID: 25787736 DOI: 10.1007/s13346-013-0135-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Strategies that utilize controlled release of drugs and proteins for tissue engineering have enormous potential to regenerate damaged organs and tissues. The multiple advantages of controlled release strategies merit overcoming the significant challenges to translation, including high costs and long, difficult regulatory pathways. This review highlights the potential of controlled release of proteins for tissue engineering and regenerative medicine. We specifically discuss treatment modalities that have reached preclinical and clinical trials, with emphasis on controlled release systems for bone tissue engineering, the most advanced application with several products already in clinic. Possible strategies to address translational and regulatory concerns are also discussed.
Collapse
Affiliation(s)
- Kara L Spiller
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street Vanderbilt Clinic 12-234, New York, NY, 10032, USA
| | | |
Collapse
|
21
|
Uchida H, Morita T, Niizuma K, Kushida Y, Kuroda Y, Wakao S, Sakata H, Matsuzaka Y, Mushiake H, Tominaga T, Borlongan CV, Dezawa M. Transplantation of Unique Subpopulation of Fibroblasts, Muse Cells, Ameliorates Experimental Stroke Possibly via Robust Neuronal Differentiation. Stem Cells 2015; 34:160-73. [DOI: 10.1002/stem.2206] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/30/2015] [Accepted: 07/12/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Hiroki Uchida
- Department of Stem Cell Biology and Histology
- Department of Neurosurgery
| | - Takahiro Morita
- Department of Stem Cell Biology and Histology
- Department of Neurosurgery
| | | | | | | | | | | | - Yoshiya Matsuzaka
- Department of Physiology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Hajime Mushiake
- Department of Physiology; Tohoku University Graduate School of Medicine; Sendai Japan
| | | | - Cesario V. Borlongan
- Department of Neurosurgery and Brain Repair; University of South Florida College of Medicine; Tampa Florida USA
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology
- Department of Anatomy and Anthropology
| |
Collapse
|
22
|
Lam J, Lowry WE, Carmichael ST, Segura T. Delivery of iPS-NPCs to the Stroke Cavity within a Hyaluronic Acid Matrix Promotes the Differentiation of Transplanted Cells. ADVANCED FUNCTIONAL MATERIALS 2014; 24:7053-7062. [PMID: 26213530 PMCID: PMC4512237 DOI: 10.1002/adfm.201401483] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Stroke is the leading cause of adult disability with ~80% being ischemic. Stem cell transplantation has been shown to improve functional recovery. However, the overall survival and differentiation of these cells is still low. The infarct cavity is an ideal location for transplantation as it is directly adjacent to the highly plastic peri-infarct region. Direct transplantation of cells near the infarct cavity has resulted in low cell viability. Here we deliver neural progenitor cells derived from induce pluripotent stem cells (iPS-NPC) to the infarct cavity of stroked mice encapsulated in a hyaluronic acid hydrogel matrix to protect the cells. To improve the overall viability of transplanted cells, each step of the transplantation process was optimized. Hydrogel mechanics and cell injection parameters were investigated to determine their effects on the inflammatory response of the brain and cell viability, respectively. Using parameters that balanced the desire to keep surgery invasiveness minimal and cell viability high, iPS-NPCs were transplanted to the stroke cavity of mice encapsulated in buffer or the hydrogel. While the hydrogel did not promote stem cell survival one week post-transplantation, it did promote differentiation of the neural progenitor cells to neuroblasts.
Collapse
Affiliation(s)
- Jonathan Lam
- University of California, Los Angeles, Biomedical Engineering
Department
| | - William E. Lowry
- University of California, Los Angeles, Department of Molecular, Cell
and Developmental Biology
- University of California, Los Angeles, Eli and Edythe Broad Center
for Regenerative Medicine
| | - S. Thomas Carmichael
- University of California, Los Angeles, Department of Neurology
- University of California, Los Angeles, David Geffen School of
Medicine
| | - Tatiana Segura
- University of California, Los Angeles, Biomedical Engineering
Department
- University of California, Los Angeles, Chemical and Biomolecular
Engineering Department
- Corresponding Author: Tatiana Segura, Department of Chemical and
Biomolecular Engineering, University of California, Los Angeles, 5531 Boelter
Hall, 420 Westwood Plaza, Los Angeles, CA 90095-1592,
, Fax: (310) 206-4107
| |
Collapse
|
23
|
Elliott Donaghue I, Tam R, Sefton MV, Shoichet MS. Cell and biomolecule delivery for tissue repair and regeneration in the central nervous system. J Control Release 2014; 190:219-27. [DOI: 10.1016/j.jconrel.2014.05.040] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/12/2014] [Accepted: 05/20/2014] [Indexed: 11/25/2022]
|
24
|
Tam RY, Fuehrmann T, Mitrousis N, Shoichet MS. Regenerative therapies for central nervous system diseases: a biomaterials approach. Neuropsychopharmacology 2014; 39:169-88. [PMID: 24002187 PMCID: PMC3857664 DOI: 10.1038/npp.2013.237] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 02/07/2023]
Abstract
The central nervous system (CNS) has a limited capacity to spontaneously regenerate following traumatic injury or disease, requiring innovative strategies to promote tissue and functional repair. Tissue regeneration strategies, such as cell and/or drug delivery, have demonstrated promising results in experimental animal models, but have been difficult to translate clinically. The efficacy of cell therapy, which involves stem cell transplantation into the CNS to replace damaged tissue, has been limited due to low cell survival and integration upon transplantation, while delivery of therapeutic molecules to the CNS using conventional methods, such as oral and intravenous administration, have been limited by diffusion across the blood-brain/spinal cord-barrier. The use of biomaterials to promote graft survival and integration as well as localized and sustained delivery of biologics to CNS injury sites is actively being pursued. This review will highlight recent advances using biomaterials as cell- and drug-delivery vehicles for CNS repair.
Collapse
Affiliation(s)
- Roger Y Tam
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada,Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Tobias Fuehrmann
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada,Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Nikolaos Mitrousis
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada,Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada,Department of Chemistry, University of Toronto, Toronto, ON, Canada,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, 160 College Street, Room 514, Toronto, ON, Canada, Tel: +416 978 1460, Fax: +416 978 4317, E-mail:
| |
Collapse
|
25
|
Huang W, Mo X, Qin C, Zheng J, Liang Z, Zhang C. Transplantation of differentiated bone marrow stromal cells promotes motor functional recovery in rats with stroke. Neurol Res 2013; 35:320-8. [PMID: 23485057 DOI: 10.1179/1743132812y.0000000151] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Wen Huang
- Department of NeurologyFirst Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xuean Mo
- Department of NeurologyFirst Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Chao Qin
- Department of NeurologyFirst Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Jinou Zheng
- Department of NeurologyFirst Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Zhijian Liang
- Department of NeurologyFirst Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Cheng Zhang
- Department of NeurologySun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Woodbury ME, Ikezu T. Fibroblast growth factor-2 signaling in neurogenesis and neurodegeneration. J Neuroimmune Pharmacol 2013; 9:92-101. [PMID: 24057103 DOI: 10.1007/s11481-013-9501-5] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 09/11/2013] [Indexed: 11/26/2022]
Abstract
Fibroblast growth factor-2 (FGF2), also known as basic FGF, is a multi-functional growth factor. One of the 22-member FGF family, it signals through receptor tyrosine kinases encoding FGFR1-4. FGF2 activates FGFRs in cooperation with heparin or heparin sulfate proteoglycan to induce its pleiotropic effects in different tissues and organs, which include potent angiogenic effects and important roles in the differentiation and function of the central nervous system (CNS). FGF2 is crucial to development of the CNS, which explains its importance in adult neurogenesis. During development, high levels of FGF2 are detected from neurulation onwards. Moreover, developmental expression of FGF2 and its receptors is temporally and spatially regulated, concurring with development of specific brain regions including the hippocampus and substantia nigra pars compacta. In adult neurogenesis, FGF2 has been implicated based on its expression and regulation of neural stem and progenitor cells in the neurogenic niches, the subventricular zone (SVZ) and the subgranular zone (SGZ) of the hippocampal dentate gyrus. FGFR1 signaling also modulates inflammatory signaling through the surface glycoprotein CD200, which regulates microglial activation. Because of its importance in adult neurogenesis and neuroinflammation, manipulation of FGF2/FGFR1 signaling has been a focus of therapeutic development for neurodegenerative disorders, such as Alzheimer's disease, multiple sclerosis, Parkinson's disease and traumatic brain injury. Novel strategies include intranasal administration of FGF2, administration of an NCAM-derived FGFR1 agonist, and chitosan-based nanoparticles for the delivery of FGF2 in pre-clinical animal models. In this review, we highlight current research towards therapeutic interventions targeting FGF2/FGFR1 in neurodegenerative disorders.
Collapse
Affiliation(s)
- Maya E Woodbury
- Graduate Program in Neuroscience, Boston University School of Medicine, Boston, MA, 02118, USA
| | | |
Collapse
|
27
|
Moshayedi P, Carmichael ST. Hyaluronan, neural stem cells and tissue reconstruction after acute ischemic stroke. BIOMATTER 2013; 3:23863. [PMID: 23507922 PMCID: PMC3732322 DOI: 10.4161/biom.23863] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Focal stroke is a disabling disease with lifelong sensory, motor and cognitive impairments. Given the paucity of effective clinical treatments, basic scientists are developing novel options for protection of the affected brain and regeneration of lost tissue. Tissue bioengineering and stem/progenitor cell treatments have both been individually pursued for stroke neural repair therapies, with some benefit in tissue recovery. Emerging directions in stroke neural repair approaches combine these two therapies to use biopolymers with stem/progenitor transplants to promote greater cell survival in the transplant and directed delivery of bioactive molecules to the transplanted cells and the adjacent injured tissue. In this review the background literature on a combined use of neural stem/progenitor cells encapsulated in hyaluronan gels is discussed and the way this therapeutic approach can affect the important processes involved in brain tissue reconstruction, such as angiogenesis, axon regeneration, neural differentiation and inflammation is clarified. The glycosaminoglycan hyaluronan can optimize those processes and be employed in a successful neural tissue engineering approach.
Collapse
Affiliation(s)
- Pouria Moshayedi
- Department of Neurology; David Geffen School of Medicine at UCLA; Los Angeles, CA USA
| | - S Thomas Carmichael
- Department of Neurology; David Geffen School of Medicine at UCLA; Los Angeles, CA USA
| |
Collapse
|
28
|
Aizawa-Kohama M, Endo T, Kitada M, Wakao S, Sumiyoshi A, Matsuse D, Kuroda Y, Morita T, Riera JJ, Kawashima R, Tominaga T, Dezawa M. Transplantation of bone marrow stromal cell-derived neural precursor cells ameliorates deficits in a rat model of complete spinal cord transection. Cell Transplant 2012; 22:1613-25. [PMID: 23127893 DOI: 10.3727/096368912x658791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
After severe spinal cord injury, spontaneous functional recovery is limited. Numerous studies have demonstrated cell transplantation as a reliable therapeutic approach. However, it remains unknown whether grafted neuronal cells could replace lost neurons and reconstruct neuronal networks in the injured spinal cord. To address this issue, we transplanted bone marrow stromal cell-derived neural progenitor cells (BM-NPCs) in a rat model of complete spinal cord transection 9 days after the injury. BM-NPCs were induced from bone marrow stromal cells (BMSCs) by gene transfer of the Notch-1 intracellular domain followed by culturing in the neurosphere method. As reported previously, BM-NPCs differentiated into neuronal cells in a highly selective manner in vitro. We assessed hind limb movements of the animals weekly for 7 weeks to monitor functional recovery after local injection of BM-NPCs to the transected site. To test the sensory recovery, we performed functional magnetic resonance imaging (fMRI) using electrical stimulation of the hind limbs. In the injured spinal cord, transplanted BM-NPCs were confirmed to express neuronal markers 7 weeks following the transplantation. Grafted cells successfully extended neurites beyond the transected portion of the spinal cord. Adjacent localization of synaptophysin and PSD-95 in the transplanted cells suggested synaptic formations. These results indicated survival and successful differentiation of BM-NPCs in the severely injured spinal cord. Importantly, rats that received BM-NPCs demonstrated significant motor recovery when compared to the vehicle injection group. Volumes of the fMRI signals in somatosensory cortex were larger in the BM-NPC-grafted animals. However, neuronal activity was diverse and not confined to the original hind limb territory in the somatosensory cortex. Therefore, reconstruction of neuronal networks was not clearly confirmed. Our results indicated BM-NPCs as an effective method to deliver neuronal lineage cells in a severely injured spinal cord. However, reestablishment of neuronal networks in completed transected spinal cord was still a challenging task.
Collapse
Affiliation(s)
- Misaki Aizawa-Kohama
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mesenchymal stem cells as therapeutic agents and potential targeted gene delivery vehicle for brain diseases. J Control Release 2012; 162:464-73. [DOI: 10.1016/j.jconrel.2012.07.034] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/23/2012] [Accepted: 07/25/2012] [Indexed: 01/01/2023]
|
30
|
Simón-yarza T, Garbayo E, Tamayo E, Prósper F, Blanco-prieto* MJ. Drug Delivery in Tissue Engineering: General Concepts. NANOSTRUCTURED BIOMATERIALS FOR OVERCOMING BIOLOGICAL BARRIERS 2012. [DOI: 10.1039/9781849735292-00501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
31
|
Abstract
Stroke remains one of the main causes of death and disability worldwide. The aging of the population is likely to result in a dramatic increase in the burden of stroke. Thus, it is not surprising that the pharmaceutical industry has invested much money in the development of pharmacotherapies for ischemic stroke. Promising experimental data, however, have almost consistently failed to produce a clinically effective neuroprotective or neurorestorative drug. Only intravenous recombinant tissue plasminogen activator (rtPA) has been approved for the treatment of acute ischemic stroke. Many pharmaceutical companies have scaled down their stroke programs and despite the unmet need, activity in the field is almost frozen. Trafermin, a recombinant form of human basic fibroblast growth factor (bFGF), is a good example of a translational failure in neuroprotection. However, trafermin may also promote neuronal plasticity after cerebral insults. Thus, clinical trials with trafermin in stroke are warranted but should be based on neuronal restoration rather than acute neuroprotection.
Collapse
|
32
|
Kitada M. Mesenchymal cell populations: development of the induction systems for Schwann cells and neuronal cells and finding the unique stem cell population. Anat Sci Int 2012; 87:24-44. [PMID: 22237924 DOI: 10.1007/s12565-011-0128-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 12/20/2011] [Indexed: 01/01/2023]
Abstract
Mesenchymal cell populations, referred to as mesenchymal stem cells or multipotent stromal cells (MSCs), which include bone marrow stromal cells (BMSCs), umbilical cord stromal cells and adipose stromal cells (ASCs), participate in tissue repair when transplanted into damaged or degenerating tissues. The trophic support and immunomodulation provided by MSCs can protect against tissue damage, and the differentiation potential of these cells may help to replace lost cells. MSCs are easily accessible and can be expanded on a large scale. In addition, BMSCs and ASCs can be harvested from the patient himself. Thus, MSCs are considered promising candidates for cell therapy. In this review, I will discuss recently discovered high-efficiency induction systems for deriving Schwann cells and neurons from MSCs. Other features of MSCs that are important for tissue repair include the self-renewing property of stem cells and their potential for differentiation. Thus, I will also discuss the stemness of MSCs and describe the discovery of a certain stem cell type among adult MSCs that can self-renew and differentiate into cells of all three germ layers. Furthermore, I will explore the prospects of using this cell population for cell therapy.
Collapse
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|