1
|
Liu D, Shen H, Zhang K, Shen Y, Wen R, He X, Long G, Li X. Functional Hydrogel Co-Remolding Migration and Differentiation Microenvironment for Severe Spinal Cord Injury Repair. Adv Healthc Mater 2024; 13:e2301662. [PMID: 37937326 DOI: 10.1002/adhm.202301662] [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: 05/31/2023] [Revised: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Spinal cord injury (SCI) activates nestin+ neural stem cells (NSCs), which can be regarded as potential seed cells for neuronal regeneration. However, the lesion microenvironment seriously hinders the migration of the nestin+ cells to the lesion epicenter and their differentiation into neurons to rebuild neural circuits. In this study, a photosensitive hydrogel scaffold is prepared as drug delivery carrier. Genetically engineered SDF1α and NT3 are designed and the scaffold is binary modified to reshape the lesion microenvironment. The binary modified scaffold can effectively induce the migration and neuronal differentiation of nestin+ NSCs in vitro. When implanted into a rat complete SCI model, many of the SCI-activated nestin+ cells migrate into the lesion site and give rise to neurons in short-term. Meanwhile, long-term repair results also show that implantation of the binary modified scaffold can effectively promote the maturation, functionalization and synaptic network reconstruction of neurons in the lesion site. In addition, animals treated with binary scaffold also showed better improvement in motor functions. The therapeutic strategy based on remolding the migration and neuronal differentiation lesion microenvironment provides a new insight into SCI repair by targeting activated nestin+ cells, which exhibits excellent clinical transformation prospects.
Collapse
Affiliation(s)
- Dingyang Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Kai Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Yeyu Shen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Runlin Wen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Xinghui He
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Ge Long
- Department of Anesthesia, the Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410078, China
| | - Xing Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| |
Collapse
|
2
|
Malekpour K, Hazrati A, Khosrojerdi A, Roshangar L, Ahmadi M. An overview to nanocellulose clinical application: Biocompatibility and opportunities in disease treatment. Regen Ther 2023; 24:630-641. [PMID: 38034858 PMCID: PMC10682839 DOI: 10.1016/j.reth.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/13/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Recently, the demand for organ transplantation has promptly increased due to the enhanced incidence of body organ failure, the increasing efficiency of transplantation, and the improvement in post-transplant outcomes. However, due to a lack of suitable organs for transplantation to fulfill current demand, significant organ shortage problems have emerged. Developing efficient technologies in combination with tissue engineering (TE) has opened new ways of producing engineered tissue substitutes. The use of natural nanoparticles (NPs) such as nanocellulose (NC) and nano-lignin should be used as suitable candidates in TE due to their desirable properties. Many studies have used these components to form scaffolds and three-dimensional (3D) cultures of cells derived from different tissues for tissue repair. Interestingly, these natural NPs can afford scaffolds a degree of control over their characteristics, such as modifying their mechanical strength and distributing bioactive compounds in a controlled manner. These bionanomaterials are produced from various sources and are highly compatible with human-derived cells as they are derived from natural components. In this review, we discuss some new studies in this field. This review summarizes the scaffolds based on NC, counting nanocrystalline cellulose and nanofibrillated cellulose. Also, the efficient approaches that can extract cellulose with high purity and increased safety are discussed. We concentrate on the most recent research on the use of NC-based scaffolds for the restoration, enhancement, or replacement of injured organs and tissues, such as cartilage, skin, arteries, brain, and bone. Finally, we suggest the experiments and promises of NC-based TE scaffolds.
Collapse
Affiliation(s)
- Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezou Khosrojerdi
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Sakti YM, Malueka RG, Dwianingsih EK, Kusumaatmaja A, Mafaza A, Emiri DM. Diamond Concept as Principle for the Development of Spinal Cord Scaffold: A Literature Review. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION: Spinal cord injury (SCI) has been bringing detrimental impacts on the affected individuals. However, not only that, it also brings a tremendous effect on the socioeconomic and health-care system. Treatment regimen and strategy for SCI patient have been under further research.
DISCUSSION: The main obstacles of regeneration on neuronal structure are the neuroinflammatory process and poor debris clearance, causing a longer healing process and an extensive inflammation process due to this particular inflammatory process. To resolve all of the mentioned significant issues in SCIs neuronal regeneration, a comprehensive model is necessary to analyze each step of progressive condition in SCI. In this review, we would like to redefine a comprehensive concept of the “Diamond Concept” from previously used in fracture management to SCI management, which consists of cellular platform, cellular inductivity, cellular conductivity, and material integrity. The scaffolding treatment strategy for SCI has been widely proposed due to its flexibility. It enables the physician to combine another treatment method such as neuroprotective or neuroregenerative or both in one intervention.
CONCLUSION: Diamond concept perspective in the implementation of scaffolding could be advantageous to increase the outcome of SCI treatment.
Collapse
|
4
|
Nelson DW, Gilbert RJ. Extracellular Matrix-Mimetic Hydrogels for Treating Neural Tissue Injury: A Focus on Fibrin, Hyaluronic Acid, and Elastin-Like Polypeptide Hydrogels. Adv Healthc Mater 2021; 10:e2101329. [PMID: 34494398 PMCID: PMC8599642 DOI: 10.1002/adhm.202101329] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/23/2021] [Indexed: 12/19/2022]
Abstract
Neurological and functional recovery is limited following central nervous system injury and severe injury to the peripheral nervous system. Extracellular matrix (ECM)-mimetic hydrogels are of particular interest as regenerative scaffolds for the injured nervous system as they provide 3D bioactive interfaces that modulate cellular response to the injury environment and provide naturally degradable scaffolding for effective tissue remodeling. In this review, three unique ECM-mimetic hydrogels used in models of neural injury are reviewed: fibrin hydrogels, which rely on a naturally occurring enzymatic gelation, hyaluronic acid hydrogels, which require chemical modification prior to chemical crosslinking, and elastin-like polypeptide (ELP) hydrogels, which exhibit a temperature-sensitive gelation. The hydrogels are reviewed by summarizing their unique biological properties, their use as drug depots, and their combination with other biomaterials, such as electrospun fibers and nanoparticles. This review is the first to focus on these three ECM-mimetic hydrogels for their use in neural tissue engineering. Additionally, this is the first review to summarize the use of ELP hydrogels for nervous system applications. ECM-mimetic hydrogels have shown great promise in preclinical models of neural injury and future advancements in their design and use can likely lead to viable treatments for patients with neural injury.
Collapse
Affiliation(s)
- Derek W Nelson
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| |
Collapse
|
5
|
Liu D, Shu M, Liu W, Shen Y, Long G, Zhao Y, Hou X, Xiao Z, Dai J, Li X. Binary scaffold facilitates in situ regeneration of axons and neurons for complete spinal cord injury repair. Biomater Sci 2021; 9:2955-2971. [PMID: 33634811 DOI: 10.1039/d0bm02212h] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The limited regrowth of transected axons and insufficient regeneration of lost neurons in adult mammals collectively hinder complete spinal cord injury (SCI) repair. Hence, designing an ideal bio-scaffold which could coordinate the regeneration of axons and neurons in situ might be able to effectively facilitate the reconstruction of neural circuits and the recovery of nerve function after complete SCI. In this study, a sponge-like collagen scaffold with good drug release characteristics and good nerve cell compatibility was prepared and used as a drug delivery platform. When doubly modified with Taxol liposomes and collagen-binding neurotrophic factor 3, the scaffold dually alleviated myelin-derived inhibition on neurite outgrowth of neurons and neuronal differentiation of neural stem cells in vitro. Meanwhile, the binary-drug modified scaffold was also able to simultaneously promote both axonal and neuronal regeneration when implanted into a complete transected SCI model. Additionally, the regenerated axons and neurons throughout the lesion site formed extensive synaptic connections. Finally, complete SCI rats that received binary scaffold implantation exhibited optimal neuroelectrophysiological recovery and hindlimb locomotor improvement. Taken together, implantation of the binary scaffold can establish neural bridging networks for functional recovery, representing a clinically promising strategy for complete SCI repair.
Collapse
Affiliation(s)
- Dingyang Liu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China.
| | - Muya Shu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yeyu Shen
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, Hunan Province, China and Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Ge Long
- Department of Anesthesia, the Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xing Li
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China. and Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, Hunan Province, China and Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
6
|
Dayekh K, Mequanint K. The effects of progenitor and differentiated cells on ectopic calcification of engineered vascular tissues. Acta Biomater 2020; 115:288-298. [PMID: 32853805 DOI: 10.1016/j.actbio.2020.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022]
Abstract
Ectopic vascular calcification associated with aging, diabetes mellitus, atherosclerosis, and chronic kidney disease is a considerable risk factor for cardiovascular events and death. Although vascular smooth muscle cells are primarily implicated in calcification, the role of progenitor cells is less known. In this study, we engineered tubular vascular tissues from embryonic multipotent mesenchymal progenitor cells either without differentiating or after differentiating them into smooth muscle cells and studied ectopic calcification through targeted gene analysis. Tissues derived from both differentiated and undifferentiated cells calcified in response to hyperphosphatemic inorganic phosphate (Pi) treatment suggesting that a single cell-type (progenitor cells or differentiated cells) may not be the sole cause of the process. We also demonstrated that Vitamin K, which is the matrix gla protein activator, had a protective role against calcification in engineered vascular tissues. Addition of partially-soluble elastin upregulated osteogenic marker genes suggesting a calcification process. Furthermore, partially-soluble elastin downregulated smooth muscle myosin heavy chain (Myh11) gene which is a late-stage differentiation marker. This latter point, in turn, suggests that SMC may be switching into a synthetic phenotype which is one feature of vascular calcification. Taken together, our approach presents a valuable tool to study ectopic calcification and associated gene expressions relevant to clinical therapeutic targets.
Collapse
|
7
|
Jiang Y, Lian XL. Heart regeneration with human pluripotent stem cells: Prospects and challenges. Bioact Mater 2020; 5:74-81. [PMID: 31989061 PMCID: PMC6965207 DOI: 10.1016/j.bioactmat.2020.01.003] [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] [Received: 09/28/2019] [Revised: 12/16/2019] [Accepted: 01/02/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular disease, ranging from congenital heart disease to adult myocardial infarction, is the leading cause of death worldwide. In pursuit of reliable cardiovascular regenerative medicine, human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer plenty of potential cell-based applications. HPSCs are capable of proliferating indefinitely in an undifferentiated state, and are also pluripotent, being able to differentiate into virtually any somatic cell types given specific stepwise cues, thus representing an unlimited source to generate functional cardiovascular cells for heart regeneration. Here we recapitulated current advances in developing efficient protocols to generate hPSC-derived cardiovascular cell lineages, including cardiomyocytes, endothelial cells, and epicardial cells. We also discussed applications of hPSC-derived cells in combination with compatible bioactive materials, promising trials of cell transplantation in animal models of myocardial infarction, and potential hurdles to bring us closer to the ultimate goal of cell-based heart repair. HPSCs hold tremendous therapeutic potential for treating CVDs. HPSCs could differentiate into multiple cardiovascular cell lineages. Transplantation of hPSC-derived cardiovascular cells and biomaterials shows promising results, but challenges still remain.
Collapse
Affiliation(s)
- Yuqian Jiang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA.,Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Xiaojun Lance Lian
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA.,Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.,Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
8
|
Xu Y, Chen C, Hellwarth PB, Bao X. Biomaterials for stem cell engineering and biomanufacturing. Bioact Mater 2019; 4:366-379. [PMID: 31872161 PMCID: PMC6909203 DOI: 10.1016/j.bioactmat.2019.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Recent years have witnessed the expansion of tissue failures and diseases. The uprising of regenerative medicine converges the sight onto stem cell-biomaterial based therapy. Tissue engineering and regenerative medicine proposes the strategy of constructing spatially, mechanically, chemically and biologically designed biomaterials for stem cells to grow and differentiate. Therefore, this paper summarized the basic properties of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. The properties of frequently used biomaterials were also described in terms of natural and synthetic origins. Particularly, the combination of stem cells and biomaterials for tissue repair applications was reviewed in terms of nervous, cardiovascular, pancreatic, hematopoietic and musculoskeletal system. Finally, stem-cell-related biomanufacturing was envisioned and the novel biofabrication technologies were discussed, enlightening a promising route for the future advancement of large-scale stem cell-biomaterial based therapeutic manufacturing.
Collapse
Affiliation(s)
| | | | | | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, West Lafayette, IN, 47907, USA
| |
Collapse
|
9
|
de la Vega L, Lee C, Sharma R, Amereh M, Willerth SM. 3D bioprinting models of neural tissues: The current state of the field and future directions. Brain Res Bull 2019; 150:240-249. [DOI: 10.1016/j.brainresbull.2019.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
|
10
|
Xing H, Ren X, Yin H, Sun C, Jiang T. Construction of a NT-3 sustained-release system cross-linked with an acellular spinal cord scaffold and its effects on differentiation of cultured bone marrow mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109902. [PMID: 31500033 DOI: 10.1016/j.msec.2019.109902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 05/28/2019] [Accepted: 06/16/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study sought to promote the adhesion, proliferation and differentiation of rat bone marrow mesenchymal stem cells by constructing a neurotrophin-3 (NT-3) sustained-release system cross-linked with an acellular spinal cord scaffold. METHODS 1-Ethyl-3-[3-dimethylaminopropyl] carbodiimide hydrochloride (EDC) chemistry combined with chemical extraction was used to construct an acellular spinal cord scaffold. The decellularization completion was validated. An EDC cross-linking method was used to construct the NT-3 cross-linked acellular spinal scaffold. ELISA was used to verify sustained release of NT-3; the dorsal root ganglion method was used to verify the biological activity of the sustained-release NT-3. DAPI staining was used to confirm the adhesion of the cultured rat bone marrow mesenchymal stem cells (P3) to the NT-3 scaffold, and cell counting kit-8 (CCK-8) analysis was used to verify the cellular proliferation after 24 h and 48 h of culture. Immunohistochemistry was used to confirm the differentiation of the bone marrow cells into neuron-like cells. RESULTS An NT-3 sustained-release system cross-linked to an acellular spinal cord scaffold was successfully constructed. Sustained-release NT-3 could persist for 35 days and had biological activity for at least 21 days. It could promote the adhesion, proliferation and differentiation of rat bone marrow mesenchymal stem cells. CONCLUSION As a composite scaffold, an NT-3 sustained-release system cross-linked with an acellular spinal cord scaffold has potential applications for tissue engineering.
Collapse
Affiliation(s)
- Hui Xing
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Xianjun Ren
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Hong Yin
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Chao Sun
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Tao Jiang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China.
| |
Collapse
|
11
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
12
|
Kornev VA, Grebenik EA, Solovieva AB, Dmitriev RI, Timashev PS. Hydrogel-assisted neuroregeneration approaches towards brain injury therapy: A state-of-the-art review. Comput Struct Biotechnol J 2018; 16:488-502. [PMID: 30455858 PMCID: PMC6232648 DOI: 10.1016/j.csbj.2018.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022] Open
Abstract
Recent years have witnessed the development of an enormous variety of hydrogel-based systems for neuroregeneration. Formed from hydrophilic polymers and comprised of up to 90% of water, these three-dimensional networks are promising tools for brain tissue regeneration. They can assist structural and functional restoration of damaged tissues by providing mechanical support and navigating cell fate. Hydrogels also show the potential for brain injury therapy due to their broadly tunable physical, chemical, and biological properties. Hydrogel polymers, which have been extensively implemented in recent brain injury repair studies, include hyaluronic acid, collagen type I, alginate, chitosan, methylcellulose, Matrigel, fibrin, gellan gum, self-assembling peptides and proteins, poly(ethylene glycol), methacrylates, and methacrylamides. When viewed as tools for neuroregeneration, hydrogels can be divided into: (1) hydrogels suitable for brain injury therapy, (2) hydrogels that do not meet basic therapeutic requirements and (3) promising hydrogels which meet the criteria for further investigations. Our analysis shows that fibrin, collagen I and self-assembling peptide-based hydrogels display very attractive properties for neuroregeneration.
Collapse
Affiliation(s)
- Vladimir A. Kornev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Ekaterina A. Grebenik
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Anna B. Solovieva
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
| | - Ruslan I. Dmitriev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
- Institute of Photonic Technologies, Research Center “Crystallography and Photonics” Russian Academy of Sciences, 2 Pionerskaya st., Troitsk, Moscow 108840, Russian Federation
| |
Collapse
|
13
|
Dalamagkas K, Tsintou M, Seifalian A, Seifalian AM. Translational Regenerative Therapies for Chronic Spinal Cord Injury. Int J Mol Sci 2018; 19:E1776. [PMID: 29914060 PMCID: PMC6032191 DOI: 10.3390/ijms19061776] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury is a chronic and debilitating neurological condition that is currently being managed symptomatically with no real therapeutic strategies available. Even though there is no consensus on the best time to start interventions, the chronic phase is definitely the most stable target in order to determine whether a therapy can effectively restore neurological function. The advancements of nanoscience and stem cell technology, combined with the powerful, novel neuroimaging modalities that have arisen can now accelerate the path of promising novel therapeutic strategies from bench to bedside. Several types of stem cells have reached up to clinical trials phase II, including adult neural stem cells, human spinal cord stem cells, olfactory ensheathing cells, autologous Schwann cells, umbilical cord blood-derived mononuclear cells, adult mesenchymal cells, and autologous bone-marrow-derived stem cells. There also have been combinations of different molecular therapies; these have been either alone or combined with supportive scaffolds with nanostructures to facilitate favorable cell⁻material interactions. The results already show promise but it will take some coordinated actions in order to develop a proper step-by-step approach to solve impactful problems with neural repair.
Collapse
Affiliation(s)
- Kyriakos Dalamagkas
- The Institute for Rehabilitation and Research, Memorial Hermann Texas Medical Centre, Houston, TX 77030, USA.
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
| | - Magdalini Tsintou
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
- Center for Neural Systems Investigations, Massachusetts General Hospital/HST Athinoula A., Martinos Centre for Biomedical Imaging, Harvard Medical School, Boston, MA 02129, USA.
| | - Amelia Seifalian
- Faculty of Medical Sciences, UCL Medical School, London WC1E 6BT, UK.
| | - Alexander M Seifalian
- NanoRegMed Ltd. (Nanotechnology & Regenerative Medicine Commercialization Centre), The London BioScience Innovation Centre, London NW1 0NH, UK.
| |
Collapse
|
14
|
Gonzalez de Torre I, Weber M, Quintanilla L, Alonso M, Jockenhoevel S, Rodríguez Cabello JC, Mela P. Hybrid elastin-like recombinamer-fibrin gels: physical characterization and in vitro evaluation for cardiovascular tissue engineering applications. Biomater Sci 2018; 4:1361-70. [PMID: 27430365 DOI: 10.1039/c6bm00300a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the field of tissue engineering, the properties of the scaffolds are of crucial importance for the success of the application. Hybrid materials combine the properties of the different components that constitute them. In this study hybrid gels of Elastin-Like Recombinamer (ELR) and fibrin were prepared with a range of polymer concentrations and ELR-to-fibrin ratios. The correlation between SEM micrographs, porosities, swelling ratios and rheological properties was discussed and a poroelastic mechanism was suggested to explain the mechanical behavior of the hybrid gels. Applicability as scaffold materials for cardiovascular tissue engineering was shown by the realization of cell-laden matrixes which supported the synthesis of collagens as revealed by immunohistochemical analysis. As a proof of concept, a tissue-engineered heart valve was fabricated by injection moulding and cultivated in a bioreactor for 3 weeks under dynamic conditions. Tissue analysis revealed the production of collagen I and III, fundamental proteins for cardiovascular constructs.
Collapse
Affiliation(s)
- Israel Gonzalez de Torre
- BIOFORGE, CIBER-BBN, Campus "Miguel Delibes" Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, 47011, Valladolid, Spain and TECHNICAL PROTEINS NANOBIOTECHNOLOGY S.L., Campus "Miguel Delibes" Edificio CTTA, Universidad de Valladolid, Paseo Belén 9A, 47011, Valladolid, Spain
| | - Miriam Weber
- Tissue Engineering and Textile Implants, AME, Helmholtz Institute, RWTH Aachen University, Pauwelsstr. 20, 52074 Aachen, Germany
| | - Luis Quintanilla
- BIOFORGE, CIBER-BBN, Campus "Miguel Delibes" Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, 47011, Valladolid, Spain
| | - Matilde Alonso
- BIOFORGE, CIBER-BBN, Campus "Miguel Delibes" Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, 47011, Valladolid, Spain
| | - Stefan Jockenhoevel
- Tissue Engineering and Textile Implants, AME, Helmholtz Institute, RWTH Aachen University, Pauwelsstr. 20, 52074 Aachen, Germany
| | - José Carlos Rodríguez Cabello
- BIOFORGE, CIBER-BBN, Campus "Miguel Delibes" Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, 47011, Valladolid, Spain
| | - Petra Mela
- Tissue Engineering and Textile Implants, AME, Helmholtz Institute, RWTH Aachen University, Pauwelsstr. 20, 52074 Aachen, Germany
| |
Collapse
|
15
|
Abstract
In this chapter we illustrate protocols to investigate growth and neurotrophic factors in human and rodent (rat and mouse)-derived embryonic stem cells. The conventional two-dimensional cell monolayer system to grow embryonic stem cells is presented, focusing on the coating strategies also using extracellular matrix components. Then, different approaches for three-dimensional stem cell culture are presented, using hydrogels and scaffolds. Quantitative polymerase chain reaction, immunocytochemistry, immunoenzymatic ELISA assay, and multiparametric assays to quantify growth and neurotrophic factor production are presented.
Collapse
|
16
|
Thomas M, Willerth SM. 3-D Bioprinting of Neural Tissue for Applications in Cell Therapy and Drug Screening. Front Bioeng Biotechnol 2017; 5:69. [PMID: 29204424 PMCID: PMC5698280 DOI: 10.3389/fbioe.2017.00069] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 10/19/2017] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative diseases affect millions of individuals in North America and cost the health-care industry billions of dollars for treatment. Current treatment options for degenerative diseases focus on physical rehabilitation or drug therapies, which temporarily mask the effects of cell damage, but quickly lose their efficacy. Cell therapies for the central nervous system remain an untapped market due to the complexity involved in growing neural tissues, controlling their differentiation, and protecting them from the hostile environment they meet upon implantation. Designing tissue constructs for the discovery of better drug treatments are also limited due to the resolution needed for an accurate cellular representation of the brain, in addition to being expensive and difficult to translate to biocompatible materials. 3-D printing offers a streamlined solution for engineering brain tissue for drug discovery or, in the future, for implantation. New microfluidic and bioplotting devices offer increased resolution, little impact on cell viability and have been tested with several bioink materials including fibrin, collagen, hyaluronic acid, poly(caprolactone), and poly(ethylene glycol). This review details current efforts at bioprinting neural tissue and highlights promising avenues for future work.
Collapse
Affiliation(s)
- Michaela Thomas
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| |
Collapse
|
17
|
Robinson M, Chapani P, Styan T, Vaidyanathan R, Willerth SM. Functionalizing Ascl1 with Novel Intracellular Protein Delivery Technology for Promoting Neuronal Differentiation of Human Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2017; 12:476-83. [PMID: 27138845 DOI: 10.1007/s12015-016-9655-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pluripotent stem cells can become any cell type found in the body. Accordingly, one of the major challenges when working with pluripotent stem cells is producing a highly homogenous population of differentiated cells, which can then be used for downstream applications such as cell therapies or drug screening. The transcription factor Ascl1 plays a key role in neural development and previous work has shown that Ascl1 overexpression using viral vectors can reprogram fibroblasts directly into neurons. Here we report on how a recombinant version of the Ascl1 protein functionalized with intracellular protein delivery technology (Ascl1-IPTD) can be used to rapidly differentiate human induced pluripotent stem cells (hiPSCs) into neurons. We first evaluated a range of Ascl1-IPTD concentrations to determine the most effective amount for generating neurons from hiPSCs cultured in serum free media. Next, we looked at the frequency of Ascl1-IPTD supplementation in the media on differentiation and found that one time supplementation is sufficient enough to trigger the neural differentiation process. Ascl1-IPTD was efficiently taken up by the hiPSCs and enabled rapid differentiation into TUJ1-positive and NeuN-positive populations with neuronal morphology after 8 days. After 12 days of culture, hiPSC-derived neurons produced by Ascl1-IPTD treatment exhibited greater neurite length and higher numbers of branch points compared to neurons derived using a standard neural progenitor differentiation protocol. This work validates Ascl1-IPTD as a powerful tool for engineering neural tissue from pluripotent stem cells.
Collapse
Affiliation(s)
- Meghan Robinson
- Biomedical Engineering Program, University of Victoria, Victoria, BC, Canada
| | - Parv Chapani
- Department of Biochemistry, University of Victoria, Victoria, BC, Canada
| | - Tara Styan
- Biomedical Engineering Program, University of Victoria, Victoria, BC, Canada
| | | | - Stephanie Michelle Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada.
| |
Collapse
|
18
|
Joshi R, Buchanan JC, Tavana H. Self-regulatory factors of embryonic stem cells in co-culture with stromal cells enhance neural differentiation. Integr Biol (Camb) 2017; 9:418-426. [PMID: 28406502 PMCID: PMC5498101 DOI: 10.1039/c7ib00038c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells (ESCs), due to their intrinsic capability to generate somatic cells of all three germ layers, are potential sources of neural cells for cell replacement therapies. However, the empirical differentiation protocols and the lack of mechanistic understanding of the neural differentiation of ESCs have limited the utility of ESCs as a developmental model or as a cell source for neural cell populations for replacement therapies. Co-culturing ESCs with stromal cells is one of the extensively used methods to induce neural differentiation. Despite several studies to identify neural inducing factors in stromal cell induced neural differentiation, the self-regulatory effects of ESCs in the neural differentiation process remain unexplored. For the first time, we elucidate the self-regulatory role of mESCs in their neural cell differentiation by supplementing conditioned media from differentiating mESCs to mESC-PA6 co-cultures and quantitatively evaluating the change in neural differentiation. Moreover, we use statistical tools to analyze the expression of various growth and trophic factors and distinguish the factors produced primarily by PA6 cells versus mESCs in co-culture. We observe that addition of the medium containing mESC-secreted factors to a single mESC colony co-cultured with PA6 cells significantly enhances the neural differentiation of mESCs compares to the medium extracted from the stromal cells only. Hierarchical clustering of gene expression data from PA6 and co-cultured mESCs segregates two groups of factors that are produced by the stromal cells and differentiating mESCs. Identifying the major soluble factors that drive and regulate the neural differentiation process in the mESC-PA6 co-culture niche will help understand molecular mechanisms of neural development. Moreover, it can be a major step toward developing novel protocols to differentiate stem cells with mESC derived factor supplementation without using feeder cells and with greater efficiency compared to existing approaches.
Collapse
Affiliation(s)
- R. Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, USA
| | - J. C. Buchanan
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, USA
| | - H. Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
19
|
Shultz RB, Wang Z, Nong J, Zhang Z, Zhong Y. Local delivery of thyroid hormone enhances oligodendrogenesis and myelination after spinal cord injury. J Neural Eng 2017; 14:036014. [PMID: 28358726 DOI: 10.1088/1741-2552/aa6450] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Traumatic spinal cord injury (SCI) causes apoptosis of myelin-forming oligodendrocytes (OLs) and demyelination of surviving axons, resulting in conduction failure. Remyelination of surviving denuded axons provides a promising therapeutic target for spinal cord repair. While cell transplantation has demonstrated efficacy in promoting remyelination and functional recovery, the lack of ideal cell sources presents a major obstacle to clinical application. The adult spinal cord contains oligodendrocyte precursor cells and multipotent neural stem/progenitor cells that have the capacity to differentiate into mature, myelinating OLs. However, endogenous oligodendrogenesis and remyelination processes are limited by the upregulation of remyelination-inhibitory molecules in the post-injury microenvironment. Multiple growth factors/molecules have been shown to promote OL differentiation and myelination. APPROACH In this study we screened these therapeutics and found that 3, 3', 5-triiodothyronine (T3) is the most effective in promoting oligodendrogenesis and OL maturation in vitro. However, systemic administration of T3 to achieve therapeutic doses in the injured spinal cord is likely to induce hyperthyroidism, resulting in serious side effects. MAIN RESULTS In this study we developed a novel hydrogel-based drug delivery system for local delivery of T3 to the injury site without eliciting systemic toxicity. SIGNIFICANCE Using a clinically relevant cervical contusion injury model, we demonstrate that local delivery of T3 at doses comparable to safe human doses promoted new mature OL formation and myelination after SCI.
Collapse
Affiliation(s)
- Robert B Shultz
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, United States of America
| | | | | | | | | |
Collapse
|
20
|
Iyer NR, Wilems TS, Sakiyama-Elbert SE. Stem cells for spinal cord injury: Strategies to inform differentiation and transplantation. Biotechnol Bioeng 2017; 114:245-259. [PMID: 27531038 PMCID: PMC5642909 DOI: 10.1002/bit.26074] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/20/2016] [Accepted: 08/07/2016] [Indexed: 12/13/2022]
Abstract
The complex pathology of spinal cord injury (SCI), involving a cascade of secondary events and the formation of inhibitory barriers, hampers regeneration across the lesion site and often results in irreversible loss of motor function. The limited regenerative capacity of endogenous cells after SCI has led to a focus on the development of cell therapies that can confer both neuroprotective and neuroregenerative benefits. Stem cells have emerged as a candidate cell source because of their ability to self-renew and differentiate into a multitude of specialized cell types. While ethical and safety concerns impeded the use of stem cells in the past, advances in isolation and differentiation methods have largely mitigated these issues. A confluence of work in stem cell biology, genetics, and developmental neurobiology has informed the directed differentiation of specific spinal cell types. After transplantation, these stem cell-derived populations can replace lost cells, provide trophic support, remyelinate surviving axons, and form relay circuits that contribute to functional recovery. Further refinement of stem cell differentiation and transplantation methods, including combinatorial strategies that involve biomaterial scaffolds and drug delivery, is critical as stem cell-based treatments enter clinical trials. Biotechnol. Bioeng. 2017;114: 245-259. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nisha R Iyer
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Thomas S Wilems
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| |
Collapse
|
21
|
Kim E, Tae G. Direct reprogramming and biomaterials for controlling cell fate. Biomater Res 2016; 20:39. [PMID: 27980804 PMCID: PMC5142385 DOI: 10.1186/s40824-016-0086-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/26/2016] [Indexed: 01/08/2023] Open
Abstract
Direct reprogramming which changes the fate of matured cell is a very useful technique with a great interest recently. This approach can eliminate the drawbacks of direct usage of stem cells and allow the patient specific treatment in regenerative medicine. Overexpression of diverse factors such as general reprogramming factors or lineage specific transcription factors can change the fate of already differentiated cells. On the other hand, biomaterials can provide physical and topographical cues or biochemical cues on cells, which can dictate or significantly affect the differentiation of stem cells. The role of biomaterials on direct reprogramming has not been elucidated much, but will be potentially significant to improve the efficiency or specificity of direct reprogramming. In this review, the strategies for general direct reprogramming and biomaterials-guided stem cell differentiation are summarized with the addition of the up-to-date progress on biomaterials for direct reprogramming.
Collapse
Affiliation(s)
- Eunsol Kim
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| |
Collapse
|
22
|
Agbay A, Edgar JM, Robinson M, Styan T, Wilson K, Schroll J, Ko J, Khadem Mohtaram N, Jun MBG, Willerth SM. Biomaterial Strategies for Delivering Stem Cells as a Treatment for Spinal Cord Injury. Cells Tissues Organs 2016; 202:42-51. [DOI: 10.1159/000446474] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2016] [Indexed: 11/19/2022] Open
Abstract
Ongoing clinical trials are evaluating the use of stem cells as a way to treat traumatic spinal cord injury (SCI). However, the inhibitory environment present in the injured spinal cord makes it challenging to achieve the survival of these cells along with desired differentiation into the appropriate phenotypes necessary to regain function. Transplanting stem cells along with an instructive biomaterial scaffold can increase cell survival and improve differentiation efficiency. This study reviews the literature discussing different types of instructive biomaterial scaffolds developed for transplanting stem cells into the injured spinal cord. We have chosen to focus specifically on biomaterial scaffolds that direct the differentiation of neural stem cells and pluripotent stem cells since they offer the most promise for producing the cell phenotypes that could restore function after SCI. In terms of biomaterial scaffolds, this article reviews the literature associated with using hydrogels made from natural biomaterials and electrospun scaffolds for differentiating stem cells into neural phenotypes. It then presents new data showing how these different types of scaffolds can be combined for neural tissue engineering applications and provides directions for future studies.
Collapse
|
23
|
Russell LN, Lampe KJ. Engineering Biomaterials to Influence Oligodendroglial Growth, Maturation, and Myelin Production. Cells Tissues Organs 2016; 202:85-101. [PMID: 27701172 DOI: 10.1159/000446645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2016] [Indexed: 11/19/2022] Open
Abstract
Millions of people suffer from damage or disease to the nervous system that results in a loss of myelin, such as through a spinal cord injury or multiple sclerosis. Diminished myelin levels lead to further cell death in which unmyelinated neurons die. In the central nervous system, a loss of myelin is especially detrimental because of its poor ability to regenerate. Cell therapies such as stem or precursor cell injection have been investigated as stem cells are able to grow and differentiate into the damaged cells; however, stem cell injection alone has been unsuccessful in many areas of neural regeneration. Therefore, researchers have begun exploring combined therapies with biomaterials that promote cell growth and differentiation while localizing cells in the injured area. The regrowth of myelinating oligodendrocytes from neural stem cells through a biomaterials approach may prove to be a beneficial strategy following the onset of demyelination. This article reviews recent advancements in biomaterial strategies for the differentiation of neural stem cells into oligodendrocytes, and presents new data indicating appropriate properties for oligodendrocyte precursor cell growth. In some cases, an increase in oligodendrocyte differentiation alongside neurons is further highlighted for functional improvements where the biomaterial was then tested for increased myelination both in vitro and in vivo.
Collapse
|
24
|
Regalado-Santiago C, Juárez-Aguilar E, Olivares-Hernández JD, Tamariz E. Mimicking Neural Stem Cell Niche by Biocompatible Substrates. Stem Cells Int 2016; 2016:1513285. [PMID: 26880934 PMCID: PMC4736764 DOI: 10.1155/2016/1513285] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/19/2015] [Accepted: 11/23/2015] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) participate in the maintenance, repair, and regeneration of the central nervous system. During development, the primary NSCs are distributed along the ventricular zone of the neural tube, while, in adults, NSCs are mainly restricted to the subependymal layer of the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus in the hippocampus. The circumscribed areas where the NSCs are located contain the secreted proteins and extracellular matrix components that conform their niche. The interplay among the niche elements and NSCs determines the balance between stemness and differentiation, quiescence, and proliferation. The understanding of niche characteristics and how they regulate NSCs activity is critical to building in vitro models that include the relevant components of the in vivo niche and to developing neuroregenerative approaches that consider the extracellular environment of NSCs. This review aims to examine both the current knowledge on neurogenic niche and how it is being used to develop biocompatible substrates for the in vitro and in vivo mimicking of extracellular NSCs conditions.
Collapse
Affiliation(s)
- Citlalli Regalado-Santiago
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Enrique Juárez-Aguilar
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Juan David Olivares-Hernández
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| |
Collapse
|
25
|
Wilems TS, Pardieck J, Iyer N, Sakiyama-Elbert SE. Combination therapy of stem cell derived neural progenitors and drug delivery of anti-inhibitory molecules for spinal cord injury. Acta Biomater 2015; 28:23-32. [PMID: 26384702 DOI: 10.1016/j.actbio.2015.09.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/28/2015] [Accepted: 09/15/2015] [Indexed: 12/31/2022]
Abstract
Regeneration of lost synaptic connections following spinal cord injury (SCI) is limited by local ischemia, cell death, and an excitotoxic environment, which leads to the development of an inhibitory glial scar surrounding a cystic cavity. While a variety of single therapy interventions provide incremental improvements to functional recovery after SCI, they are limited; a multifactorial approach that combines several single therapies may provide a better chance of overcoming the multitude of obstacles to recovery. To this end, fibrin scaffolds were modified to provide sustained delivery of neurotrophic factors and anti-inhibitory molecules, as well as encapsulation of embryonic stem cell-derived progenitor motor neurons (pMNs). In vitro characterization of this combination scaffold confirmed that pMN viability was unaffected by culture alongside sustained delivery systems. When transplanted into a rat sub-acute SCI model, fibrin scaffolds containing growth factors (GFs), anti-inhibitory molecules without pMNs, or pMNs with GFs had lower chondroitin sulfate proteoglycan levels compared to scaffolds containing anti-inhibitory molecules with pMNs. Scaffolds containing pMNs, but not anti-inhibitory molecules, showed survival, differentiation into neuronal cell types, axonal extension in the transplant area, and the ability to integrate into host tissue. However, the combination of pMNs with sustained-delivery of anti-inhibitory molecules led to reduced cell survival and increased macrophage infiltration. While combination therapies retain potential for effective treatment of SCI, further work is needed to improve cell survival and to limit inflammation. STATEMENT OF SIGNIFICANCE Spinal cord injury (SCI) creates a highly complex inhibitory environment with a multitude of obstacles that limit recovery. Many therapeutic options have been developed to overcome single obstacles, but single therapies typically only lead to limited functional improvement. Therefore combination therapies may improve recovery by targeting several inhibitory obstacles simultaneously. The present study used biomaterial scaffolds to combine the sustained release of anti-inhibitory molecules and growth factors with cell transplantation of highly purified progenitor motor neurons. This expands upon previously established biomaterial scaffolds by supporting surviving cells, limiting inhibition from the extracellular environment, and replenishing lost cell populations. We show that while promising, certain combinations may exacerbate negative side-effects instead of augmenting positive features.
Collapse
|
26
|
Elliott Donaghue I, Shoichet MS. Controlled release of bioactive PDGF-AA from a hydrogel/nanoparticle composite. Acta Biomater 2015; 25:35-42. [PMID: 26257128 DOI: 10.1016/j.actbio.2015.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/28/2015] [Accepted: 08/05/2015] [Indexed: 12/23/2022]
Abstract
Polymer excipients, such as low molar mass poly(ethylene glycol) (PEG), have shown contradictory effects on protein stability when co-encapsulated in polymeric nanoparticles. To gain further insight into these effects, platelet-derived growth factor (PDGF-AA) was encapsulated in polymeric nanoparticles with vs. without PEG. PDGF-AA is a particularly compelling protein, as it has been demonstrated to promote cell survival and induce the oligodendrocyte differentiation of neural stem/progenitor cells (NSPCs) both in vitro and in vivo. Here we show, for the first time, the controlled release of bioactive PDGF-AA from an injectable nanoparticle/hydrogel drug delivery system (DDS). PDGF-AA was encapsulated, with high efficiency, in poly(lactide-co-glycolide) nanoparticles, and its release from the drug delivery system was followed over 21 d. Interestingly, the co-encapsulation of low molecular weight poly(ethylene glycol) increased the PDGF-AA loading but, unexpectedly, accelerated the aggregation of PDGF-AA, resulting in reduced activity and detection by enzyme-linked immunosorbent assay (ELISA). In the absence of PEG, released PDGF-AA remained bioactive as demonstrated with NSPC oligodendrocyte differentiation, similar to positive controls, and significantly different from untreated controls. This work presents a novel delivery method for differentiation factors, such as PDGF-AA, and provides insights into the contradictory effects reported in the literature of excipients, such as PEG, on the loading and release of proteins from polymeric nanoparticles. STATEMENT OF SIGNIFICANCE Previously, the polymer poly(ethylene glycol) (PEG) has been used in many biomaterials applications, from surface coatings to the encapsulation of proteins. In this work, we demonstrate that, unexpectedly, low molecular weight PEG has a deleterious effect on the release of the encapsulated protein platelet-derived growth factor AA (PDGF-AA). We also demonstrate release of bioactive PDGF-AA (in the absence of PEG). Specifically, we demonstrate the differentiation of neural stem and progenitor cells to oligodendrocytes, similar to what is observed with the addition of fresh PDGFAA. A differentiated oligodendrocyte population is a key strategy in central nervous system regeneration. This work is the first demonstration of controlled PDGF-AA release, and also brings new insights to the broader field of protein encapsulation.
Collapse
Affiliation(s)
- Irja Elliott Donaghue
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada; Department of Chemistry, University of Toronto, Canada.
| |
Collapse
|
27
|
Gomez JC, Edgar JM, Agbay AM, Bibault E, Montgomery A, Mohtaram NK, Willerth SM. Incorporation of Retinoic Acid Releasing Microspheres into Pluripotent Stem Cell Aggregates for Inducing Neuronal Differentiation. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0401-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
28
|
Affect of antidepressants on the in vitro differentiation of rat bone marrow mesenchymal stem cells into neuronal cells. Eur J Pharm Sci 2015; 73:81-7. [DOI: 10.1016/j.ejps.2015.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 01/27/2015] [Accepted: 03/23/2015] [Indexed: 12/21/2022]
|
29
|
Tara S, Krishnan LK. Bioengineered fibrin-based niche to direct outgrowth of circulating progenitors into neuron-like cells for potential use in cellular therapy. J Neural Eng 2015; 12:036011. [PMID: 25946462 DOI: 10.1088/1741-2560/12/3/036011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Autologous cells are considered to be the best choice for use in transplantation therapy. However, the challenges and risks associated with the harvest of transplantable autologous cells limit their successful therapeutic application. The current study explores the possibility of isolating neural progenitor cells from circulating multipotent adult progenitor cells for potential use in cell-based and patient-specific therapy for neurological diseases. APPROACH To enable the selection of neural progenitor cells from human peripheral blood mononuclear cells, and to support their lineage maintenance, the composition of a fibrin-based niche was optimized. Morphological examination and specific marker analysis were carried out, employing a qualitative/quantitative polymerase chain reaction followed by immunocytochemistry to: (i) characterize neural progenitor cells in culture; (ii) monitor proliferation/survival; and (iii) track their differentiation status. MAIN RESULTS The presence of neural progenitors in circulation was confirmed by the presence of nestin(+) cells at the commencement of the culture. The isolation, proliferation and differentiation of circulating neural progenitors to neuron-like cells were directed by the engineered niche. Neural cell isolation to near homogeneity was confirmed by the expression of β-III tubulin in ∼95% of cells, whereas microtubule associated protein-2 expression confirmed their ability to differentiate. The concentration of potassium chloride in the niche was found to favour neuron-like cell lengthening, cell-cell contact, and expressions of synaptophysin and tyrosine hydroxylase. SIGNIFICANCE The purpose of this research was to find out if peripheral blood could serve as a potential source of neural progenitors for cell based therapy. The study established that neural progenitors could be selectively isolated from peripheral blood mononuclear cells using a biomimetic niche. The selected cells could multiply and slowly differentiate into neuron-like cells. These neuron-like cells expressed functional proteins-tyrosine hydroxylase and synaptophysin. Early progenitors that proliferate while expressing β-III tubulin could be harvested from the culture, suggesting their potential use in cell transplantation therapy.
Collapse
Affiliation(s)
- S Tara
- Thrombosis Research Unit, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India 695012
| | | |
Collapse
|
30
|
Robinson M, Yau SY, Sun L, Gabers N, Bibault E, Christie BR, Willerth SM. Optimizing Differentiation Protocols for Producing Dopaminergic Neurons from Human Induced Pluripotent Stem Cells for Tissue Engineering Applications: Supplementary Issue: Stem Cell Biology. Biomark Insights 2015; 10 Suppl 1:61-70. [PMID: 36876191 PMCID: PMC9980910 DOI: 10.4137/bmi.s20064] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/02/2015] [Accepted: 03/05/2015] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that results when the dopaminergic neurons (DNs) present in the substantia nigra necessary for voluntary motor control are depleted, making patients with this disorder ideal candidates for cell replacement therapy. Human induced pluripotent stem cells (hiPSCs), obtained by reprogramming adult cells, possess the properties of pluripotency and immortality while enabling the possibility of patient-specific therapies. An effective cell therapy for PD requires an efficient, defined method of DN generation, as well as protection from the neuroinflammatory environment upon engraftment. Although similar in pluripotency to human embryonic stem cells (hESCs), hiPSCs differentiate less efficiently into neuronal subtypes. Previous work has shown that treatment with guggulsterone can efficiently differentiate hESCs into DNs. Our work shows that guggulsterone is able to derive DNs from hiPSCs with comparable efficiency, and furthermore, this differentiation can be achieved inside three-dimensional fibrin scaffolds that could enhance cell survival upon engraftment.
Collapse
Affiliation(s)
| | - Suk-Yu Yau
- Department of Neuroscience, Division of Medical Sciences
| | - Lin Sun
- Department of Neuroscience, Division of Medical Sciences
| | - Nicole Gabers
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | | | - Stephanie M Willerth
- Department of Biomedical Engineering.,Department of Neuroscience, Division of Medical Sciences.,Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
31
|
Mohtaram NK, Ko J, King C, Sun L, Muller N, Jun MBG, Willerth SM. Electrospun biomaterial scaffolds with varied topographies for neuronal differentiation of human-induced pluripotent stem cells. J Biomed Mater Res A 2014; 103:2591-601. [PMID: 25524598 DOI: 10.1002/jbm.a.35392] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 12/12/2014] [Indexed: 11/09/2022]
Abstract
In this study, we investigated the effect of micro and nanoscale scaffold topography on promoting neuronal differentiation of human induced pluripotent stem cells (iPSCs) and directing the resulting neuronal outgrowth in an organized manner. We used melt electrospinning to fabricate poly (ε-caprolactone) (PCL) scaffolds with loop mesh and biaxial aligned microscale topographies. Biaxial aligned microscale scaffolds were further functionalized with retinoic acid releasing PCL nanofibers using solution electrospinning. These scaffolds were then seeded with neural progenitors derived from human iPSCs. We found that smaller diameter loop mesh scaffolds (43.7 ± 3.9 µm) induced higher expression of the neural markers Nestin and Pax6 compared to thicker diameter loop mesh scaffolds (85 ± 4 µm). The loop mesh and biaxial aligned scaffolds guided the neurite outgrowth of human iPSCs along the topographical features with the maximum neurite length of these cells being longer on the biaxial aligned scaffolds. Finally, our novel bimodal scaffolds also supported the neuronal differentiation of human iPSCs as they presented both physical and chemical cues to these cells, encouraging their differentiation. These results give insight into how physical and chemical cues can be used to engineer neural tissue.
Collapse
Affiliation(s)
- Nima Khadem Mohtaram
- Department of Mechanical Engineering, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Junghyuk Ko
- Department of Mechanical Engineering, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Craig King
- Department of Biomedical Engineering, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Lin Sun
- Division of Medical Sciences, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Nathan Muller
- Department of Mechanical Engineering, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Martin Byung-Guk Jun
- Department of Mechanical Engineering, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada.,Division of Medical Sciences, University of Victoria, STN CSC, Victoria, British Columbia, V8W 2Y2, Canada.,International Collaboration on Repair Discoveries (ICORD), Vancouver, British Columbia, V5Z 1M9, Canada
| |
Collapse
|
32
|
Montgomery A, Wong A, Gabers N, Willerth SM. Engineering personalized neural tissue by combining induced pluripotent stem cells with fibrin scaffolds. Biomater Sci 2014. [PMID: 26218131 DOI: 10.1039/c4bm00299g] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are generated from adult somatic cells through the induction of key transcription factors that restore the ability to become any cell type found in the body. These cells are of interest for tissue engineering due to their potential for developing patient-specific therapies. As the technology for generating iPSCs advances, it is important to concurrently investigate protocols for the efficient differentiation of these cells to desired downstream phenotypes in combination with biomaterial scaffolds as a way of engineering neural tissue. For such applications, the generation of neurons within three dimensional fibrin scaffolds has been well characterized as a cell-delivery platform for murine embryonic stem cells (ESCs) but has not yet been applied to murine iPSCs. Given that iPSCs have been reported to differentiate less effectively than ESCs, a key objective of this investigation is to maximize the proportion of iPSC-derived neurons in fibrin through the choice of differentiation protocol. To this end, this study compares two EB-mediated protocols for generating neurons from murine iPSCs and ESCs: an 8 day 4-/4+ protocol using soluble retinoic acid in the last 4 days and a 6 day 2-/4+ protocol using soluble retinoic acid and the small molecule sonic hedgehog agonist purmorphamine in the last 4 days. EBs were then seeded in fibrin scaffolds for 14 days to allow further differentiation into neurons. EBs generated by the 2-/4+ protocol yielded a higher percentage of neurons compared to those from the 4-/4+ protocol for both iPSCs and ESCs. The results demonstrate the successful translation of the fibrin-based cell-delivery platform for use with murine iPSCs and furthermore that the proportion of neurons generated from murine iPSC-derived EBs seeded in fibrin can be maximized using the 2-/4+ differentiation protocol. Together, these findings validate the further exploration of 3D fibrin-based scaffolds as a method of delivering neuronal cells derived from iPSCs - an important step toward the development of iPSC-based tissue engineering strategies for spinal cord injury repair.
Collapse
Affiliation(s)
- Amy Montgomery
- Department of Mechanical Engineering, University of Victoria, Canada.
| | | | | | | |
Collapse
|
33
|
Adult-Derived Pluripotent Stem Cells. World Neurosurg 2014; 82:500-8. [DOI: 10.1016/j.wneu.2013.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 08/07/2013] [Accepted: 08/09/2013] [Indexed: 01/27/2023]
|
34
|
Madigan NN, Chen BK, Knight AM, Rooney GE, Sweeney E, Kinnavane L, Yaszemski MJ, Dockery P, O'Brien T, McMahon SS, Windebank AJ. Comparison of cellular architecture, axonal growth, and blood vessel formation through cell-loaded polymer scaffolds in the transected rat spinal cord. Tissue Eng Part A 2014; 20:2985-97. [PMID: 24854680 DOI: 10.1089/ten.tea.2013.0551] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The use of multichannel polymer scaffolds in a complete spinal cord transection injury serves as a deconstructed model that allows for control of individual variables and direct observation of their effects on regeneration. In this study, scaffolds fabricated from positively charged oligo[poly(ethylene glycol)fumarate] (OPF(+)) hydrogel were implanted into rat spinal cords following T9 complete transection. OPF(+) scaffold channels were loaded with either syngeneic Schwann cells or mesenchymal stem cells derived from enhanced green fluorescent protein transgenic rats (eGFP-MSCs). Control scaffolds contained extracellular matrix only. The capacity of each scaffold type to influence the architecture of regenerated tissue after 4 weeks was examined by detailed immunohistochemistry and stereology. Astrocytosis was observed in a circumferential peripheral channel compartment. A structurally separate channel core contained scattered astrocytes, eGFP-MSCs, blood vessels, and regenerating axons. Cells double-staining with glial fibrillary acid protein (GFAP) and S-100 antibodies populated each scaffold type, demonstrating migration of an immature cell phenotype into the scaffold from the animal. eGFP-MSCs were distributed in close association with blood vessels. Axon regeneration was augmented by Schwann cell implantation, while eGFP-MSCs did not support axon growth. Methods of unbiased stereology provided physiologic estimates of blood vessel volume, length and surface area, mean vessel diameter, and cross-sectional area in each scaffold type. Schwann cell scaffolds had high numbers of small, densely packed vessels within the channels. eGFP-MSC scaffolds contained fewer, larger vessels. There was a positive linear correlation between axon counts and vessel length density, surface density, and volume fraction. Increased axon number also correlated with decreasing vessel diameter, implicating the importance of blood flow rate. Radial diffusion distances in vessels significantly correlated to axon number as a hyperbolic function, showing a need to engineer high numbers of small vessels in parallel to improving axonal densities. In conclusion, Schwann cells and eGFP-MSCs influenced the regenerating microenvironment with lasting effect on axonal and blood vessel growth. OPF(+) scaffolds in a complete transection model allowed for a detailed comparative, histologic analysis of the cellular architecture in response to each cell type and provided insight into physiologic characteristics that may support axon regeneration.
Collapse
Affiliation(s)
- Nicolas N Madigan
- 1 Department of Neurology, Mayo Clinic College of Medicine , Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lu P, Graham L, Wang Y, Wu D, Tuszynski M. Promotion of survival and differentiation of neural stem cells with fibrin and growth factor cocktails after severe spinal cord injury. J Vis Exp 2014:e50641. [PMID: 25145787 DOI: 10.3791/50641] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Neural stem cells (NSCs) can self-renew and differentiate into neurons and glia. Transplanted NSCs can replace lost neurons and glia after spinal cord injury (SCI), and can form functional relays to re-connect spinal cord segments above and below a lesion. Previous studies grafting neural stem cells have been limited by incomplete graft survival within the spinal cord lesion cavity. Further, tracking of graft cell survival, differentiation, and process extension had not been optimized. Finally, in previous studies, cultured rat NSCs were typically reported to differentiate into glia when grafted to the injured spinal cord, rather than neurons, unless fate was driven to a specific cell type. To address these issues, we developed new methods to improve the survival, integration and differentiation of NSCs to sites of even severe SCI. NSCs were freshly isolated from embryonic day 14 spinal cord (E14) from a stable transgenic Fischer 344 rat line expressing green fluorescent protein (GFP) and were embedded into a fibrin matrix containing growth factors; this formulation aimed to retain grafted cells in the lesion cavity and support cell survival. NSCs in the fibrin/growth factor cocktail were implanted two weeks after thoracic level-3 (T3) complete spinal cord transections, thereby avoiding peak periods of inflammation. Resulting grafts completely filled the lesion cavity and differentiated into both neurons, which extended axons into the host spinal cord over remarkably long distances, and glia. Grafts of cultured human NSCs expressing GFP resulted in similar findings. Thus, methods are defined for improving neural stem cell grafting, survival and analysis of in vivo findings.
Collapse
Affiliation(s)
- Paul Lu
- Veterans Administration Medical Center, San Diego; Department of Neurosciences, University of California, San Diego;
| | - Lori Graham
- Department of Neurosciences, University of California, San Diego
| | - Yaozhi Wang
- Department of Neurosciences, University of California, San Diego
| | - Di Wu
- Department of Neurosciences, University of California, San Diego
| | - Mark Tuszynski
- Veterans Administration Medical Center, San Diego; Department of Neurosciences, University of California, San Diego
| |
Collapse
|
36
|
Riopel M, Trinder M, Wang R. Fibrin, a scaffold material for islet transplantation and pancreatic endocrine tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:34-44. [PMID: 24947304 DOI: 10.1089/ten.teb.2014.0188] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Fibrin is derived from fibrinogen during injury to produce a blood clot and thus promote wound repair. Composed of different domains, including Arg-Gly-Asp amino acid motifs, fibrin is used extensively as a hydrogel and sealant in the clinic. By binding to cell surface receptors like integrins and acting as a supportive 3D scaffold, fibrin has been useful in promoting cell differentiation, proliferation, function, and survival. In particular, fibrin has been able to maintain islet cell architecture, promote beta cell insulin secretion, and islet angiogenesis, as well as inducing a protective effect against cell death. During islet transplantation, fibrin improved neovascularization and islet function. These improvements resulted in reduced number of transplanted islets necessary to reverse diabetes. Therefore, fibrin, as a biocompatible and biodegradable scaffold, should be considered during subcutaneous islet transplantation and beta cell expansion in vitro to ensure maintenance of islet cell function, proliferation, and survival to develop effective cell-based therapies for the treatment of diabetes.
Collapse
Affiliation(s)
- Matthew Riopel
- 1 Children's Health Research Institute, London, Ontario, Canada
| | | | | |
Collapse
|
37
|
Task K, D'Amore A, Singh S, Candiello J, Jaramillo M, Wagner WR, Kumta P, Banerjee I. Systems level approach reveals the correlation of endoderm differentiation of mouse embryonic stem cells with specific microstructural cues of fibrin gels. J R Soc Interface 2014; 11:20140009. [PMID: 24718448 DOI: 10.1098/rsif.2014.0009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells receive numerous cues from their associated substrate that help to govern their behaviour. However, identification of influential substrate characteristics poses difficulties because of their complex nature. In this study, we developed an integrated experimental and systems level modelling approach to investigate and identify specific substrate features influencing differentiation of mouse embryonic stem cells (mESCs) on a model fibrous substrate, fibrin. We synthesized a range of fibrin gels by varying fibrinogen and thrombin concentrations, which led to a range of substrate stiffness and microstructure. mESCs were cultured on each of these gels, and characterization of the differentiated cells revealed a strong influence of substrate modulation on gene expression patterning. To identify specific substrate features influencing differentiation, the substrate microstructure was quantified by image analysis and correlated with stem cell gene expression patterns using a statistical model. Significant correlations were observed between differentiation and microstructure features, specifically fibre alignment. Furthermore, this relationship occurred in a lineage-specific manner towards endoderm. This systems level approach allows for identification of specific substrate features from a complex material which are influential to cellular behaviour. Such analysis may be effective in guiding the design of scaffolds with specific properties for tissue engineering applications.
Collapse
Affiliation(s)
- Keith Task
- Department of Chemical Engineering, University of Pittsburgh, , Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Dorj B, Won JE, Purevdorj O, Patel KD, Kim JH, Lee EJ, Kim HW. A novel therapeutic design of microporous-structured biopolymer scaffolds for drug loading and delivery. Acta Biomater 2014; 10:1238-50. [PMID: 24239677 DOI: 10.1016/j.actbio.2013.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 11/05/2013] [Accepted: 11/06/2013] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3-D) open-channeled scaffolds of biopolymers are a promising candidate matrix for tissue engineering. When scaffolds have the capacity to deliver bioactive molecules the potential for tissue regeneration should be greatly enhanced. In order to improve drug-delivery capacity, we exploit 3-D poly(lactic acid) (PLA) scaffolds by creating microporosity within the scaffold network. Macroporous channeled PLA with a controlled pore configuration was obtained by a robotic dispensing technique. In particular, a room temperature ionic liquid (RTIL) bearing hydrophilic counter-anions, such as OTf and Cl, was introduced to the biopolymer solution at varying ratios. The RTIL-biopolymer slurry was homogenized by ultrasonication, and then solidified through the robotic dispensing process, during which the biopolymer and RTIL formed a bicontinuous interpenetrating network. After ethanol wash-out treatment the RTIL was completely removed to leave highly microporous open channels throughout the PLA network. The resultant pore size was observed to be a few micrometers (average 2.43 μm) and microporosity was determined to be ∼ 70%. The microporous surface was also shown to favor initial cell adhesion, stimulating cell anchorage on the microporous structure. Furthermore, in vivo tissue responses assessed in rat subcutaneous tissue revealed good tissue compatibility, with minimal inflammatory reactions, while gathering a larger population of fibroblastic cells than the non-microporous scaffolds, and even facilitating invasion of the cells within the microporous structure. The efficacy of the micropore networks generated within the 3-D scaffolds in loading and releasing therapeutic molecules was addressed using antibiotic sodium ampicillin and protein cytochrome C as model drugs. The microporous scaffolds exhibited significantly enhanced drug loading capacity: 4-5 times increase in ampicillin and 9-10 times increase in cytochrome C compared to the non-microporous scaffolds. The release of ampicillin loaded within the microporous scaffolds was initially fast (∼ 85% for 1 week), and was then slowed down, showing a continual release up to a month. On the other hand, cytochrome C was shown to release in a highly sustainable manner over a month, without showing an initial burst release effect. This study provides a novel insight into the generation of 3-D biopolymer scaffolds with high performance in loading and delivery of biomolecules, facilitated by the creation of microporous channels through the scaffold network. The capacity to support tissue cells while in situ delivering drug molecules makes the current scaffolds potentially useful for therapeutic tissue engineering.
Collapse
|
39
|
McCreedy DA, Wilems TS, Xu H, Butts JC, Brown CR, Smith AW, Sakiyama-Elbert SE. Survival, Differentiation, and Migration of High-Purity Mouse Embryonic Stem Cell-derived Progenitor Motor Neurons in Fibrin Scaffolds after Sub-Acute Spinal Cord Injury. Biomater Sci 2014; 2:1672-1682. [PMID: 25346848 DOI: 10.1039/c4bm00106k] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Embryonic stem (ES) cells can be differentiated into many neural cell types that hold great potential as cell replacement therapies following spinal cord injury (SCI). Coupling stem cell transplantation with biomaterial scaffolds can produce a unified combination therapy with several potential advantages including enhanced cell survival, greater transplant retention, reduced scarring, and improved integration at the transplant/host interface. Undesired cell types, however, are commonly present in ES-cell derived cultures due to the limited efficiency of most ES cell induction protocols. Heterogeneous cell populations can confound the interaction between the biomaterial and specific neural populations leading to undesired outcomes. In particular, biomaterials scaffolds may enhance tumor formation by promoting survival and proliferation of undifferentiated ES cells that can persist after induction. Methods for purification of specific ES cell-derived neural populations are necessary to recognize the full potential of combination therapies involving biomaterials and ES cell-derived neural populations. We previously developed a method for enriching ES cell-derived progenitor motor neurons (pMNs) induced from mouse ES cells via antibiotic selection and showed that the enriched cell populations are depleted of pluripotent stem cells. In this study, we demonstrate the survival and differentiation of enriched pMNs within three dimensional (3D) fibrin scaffolds in vitro and when transplanted into a sub-acute dorsal hemisection model of SCI into neurons, oligodendrocytes and astrocytes.
Collapse
Affiliation(s)
- D A McCreedy
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - T S Wilems
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - H Xu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - J C Butts
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - C R Brown
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - A W Smith
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - S E Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| |
Collapse
|
40
|
Partial restoration of cardiovascular function by embryonic neural stem cell grafts after complete spinal cord transection. J Neurosci 2013; 33:17138-49. [PMID: 24155317 DOI: 10.1523/jneurosci.2851-13.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
High-level spinal cord injury can lead to cardiovascular dysfunction, including disordered hemodynamics at rest and autonomic dysreflexia during noxious stimulation. To restore supraspinal control of sympathetic preganglionic neurons (SPNs), we grafted embryonic brainstem-derived neural stem cells (BS-NSCs) or spinal cord-derived neural stem cells (SC-NSCs) expressing green fluorescent protein into the T4 complete transection site of adult rats. Animals with injury alone served as controls. Implanting of BS-NSCs but not SC-NSCs resulted in recovery of basal cardiovascular parameters, whereas both cell grafts alleviated autonomic dysreflexia. Subsequent spinal cord retransection above the graft abolished the recovery of basal hemodynamics and reflexic response. BS-NSC graft-derived catecholaminergic and serotonergic neurons showed remarkable long-distance axon growth and topographical innervation of caudal SPNs. Anterograde tracing indicated growth of medullar axons into stem cell grafts and formation of synapses. Thus, grafted embryonic brainstem-derived neurons can act as functional relays to restore supraspinal regulation of denervated SPNs, thereby contributing to cardiovascular functional improvement.
Collapse
|
41
|
Davari M, Soheili ZS, Ahmadieh H, Sanie-Jahromi F, Ghaderi S, Kanavi MR, Samiei S, Akrami H, Haghighi M, Javidi-Azad F. Amniotic fluid promotes the appearance of neural retinal progenitors and neurons in human RPE cell cultures. Mol Vis 2013; 19:2330-42. [PMID: 24265548 PMCID: PMC3834594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 11/15/2013] [Indexed: 10/25/2022] Open
Abstract
PURPOSE Retinal pigment epithelial (RPE) cells are capable of differentiating into retinal neurons when induced by the appropriate growth factors. Amniotic fluid contains a variety of growth factors that are crucial for the development of a fetus. In this study, the effects of human amniotic fluid (HAF) on primary RPE cell cultures were evaluated. METHODS RPE cells were isolated from the globes of postnatal human cadavers. The isolated cells were plated and grown in DMEM/F12 with 10% fetal bovine serum. To confirm the RPE identity of the cultured cells, they were immunocytochemically examined for the presence of the RPE cell-specific marker RPE65. RPE cultures obtained from passages 2-7 were treated with HAF and examined morphologically for 1 month. To determine whether retinal neurons or progenitors developed in the treated cultures, specific markers for bipolar (protein kinase C isomer α, PKCα), amacrine (cellular retinoic acid-binding protein I, CRABPI), and neural progenitor (NESTIN) cells were sought, and the amount of mRNA was quantified using real-time PCR. RESULTS Treating RPE cells with HAF led to a significant decrease in the number of RPE65-positive cells, while PKCα- and CRABPI-positive cells were detected in the cultures. Compared with the fetal bovine serum-treated cultures, the levels of mRNAs quantitatively increased by 2-, 20- and 22-fold for NESTIN, PKCα, and CRABPI, respectively. The RPE cultures treated with HAF established spheres containing both pigmented and nonpigmented cells, which expressed neural progenitor markers such as NESTIN. CONCLUSIONS This study showed that HAF can induce RPE cells to transdifferentiate into retinal neurons and progenitor cells, and that it provides a potential source for cell-based therapies to treat retinal diseases.
Collapse
Affiliation(s)
- Maliheh Davari
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Hamid Ahmadieh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shima Ghaderi
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Samiei
- Iranian Blood Transfusion Organization Research Center, Tehran, Iran
| | - Hassan Akrami
- Department of Biology, School of Science, Razi University, Kermanshah, Iran
| | | | | |
Collapse
|
42
|
Prabhakaran MP, Mobarakeh LG, Kai D, Karbalaie K, Nasr-Esfahani MH, Ramakrishna S. Differentiation of embryonic stem cells to cardiomyocytes on electrospun nanofibrous substrates. J Biomed Mater Res B Appl Biomater 2013; 102:447-54. [PMID: 24039141 DOI: 10.1002/jbm.b.33022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 06/24/2013] [Accepted: 08/01/2013] [Indexed: 12/31/2022]
Abstract
The potential of pluripotent embryonic stem cells (ESCs) isolated from the inner mass of blastocysts are investigated for its ability to differentiate on biocompatible electrospun nanofibers, for regeneration of the myocardially infracted heart. Nanostructured poly(d,l-lactide-co-glycolide)/collagen (PLGA/Col) scaffolds with fiber diameters in the range of 300 ± 65 nm, was fabricated by electrospinning to mimic the extracellular matrix of the native tissue. During the culture of embryoid bodies outgrowth on the scaffolds, and further differentiation of ESCs to cardiomyocytes, the PLGA/Col nanofibers was found better than that of the electrospun PLGA nanofibers, where a better interaction and growth of ESC differentiated cardiomyocytes was observed on the composite scaffolds. The phenotypical characteristics of ESC-derived cardiomyocytes and molecular protein expression were carried out by scanning electron microscopy and immunocytochemistry, respectively. Our studies highlight the significance of a suitable material, its architecture, and cell-biomaterial interactions that is essential at a nanoscale level signifying the application of a bioengineered cardiac graft for stem cell differentiation and transplantation, which could be an intriguing strategy for cardiac regeneration.
Collapse
Affiliation(s)
- Molamma P Prabhakaran
- Center for Nanofibers and Nanotechnology, E3-05-14, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, Singapore, 117576
| | | | | | | | | | | |
Collapse
|
43
|
Ko J, Mohtaram NK, Ahmed F, Montgomery A, Carlson M, Lee PC, Willerth SM, Jun MB. Fabrication of poly (ϵ-caprolactone) microfiber scaffolds with varying topography and mechanical properties for stem cell-based tissue engineering applications. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2013; 25:1-17. [DOI: 10.1080/09205063.2013.830913] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
44
|
Rehmann MS, Kloxin AM. Tunable and dynamic soft materials for three-dimensional cell culture. SOFT MATTER 2013; 9:6737-6746. [PMID: 23930136 PMCID: PMC3733394 DOI: 10.1039/c3sm50217a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/01/2013] [Indexed: 05/14/2023]
Abstract
The human body is complex and hierarchically structured, composed of cells residing within the extracellular matrix (ECM) of tissues that are assembled into organs, all working together to complete a given function. One goal of current biomaterials research is to capture some of this complexity outside of the body for understanding the underlying biology of development, repair, and disease and to devise new strategies for regenerative medicine or disease treatment. Polymeric materials have arisen as powerful tools to mimic the native ECM, giving experimenters a way to capture key aspects of the native cellular environment outside of the body. In particular, dynamic materials allow changes in the properties of these ECM mimics during an experiment, affording an additional degree of control for the experimenter. In this tutorial review, the basic cellular processes of cell migration, proliferation, and differentiation will be overviewed to motivate design considerations for polymeric ECM mimics, and examples will be given of how classes of dynamic materials are being used to study each cellular process.
Collapse
Affiliation(s)
- Matthew S. Rehmann
- Department of Chemical & Biomolecular Engineering , University of Delaware , Newark , DE 19716 , USA .
| | - April M. Kloxin
- Department of Chemical & Biomolecular Engineering , University of Delaware , Newark , DE 19716 , USA .
- Department of Materials Science & Engineering , University of Delaware , Newark , DE 19716 , USA
| |
Collapse
|
45
|
Smith AW, Hoyne JD, Nguyen PK, McCreedy DA, Aly H, Efimov IR, Rentschler S, Elbert DL. Direct reprogramming of mouse fibroblasts to cardiomyocyte-like cells using Yamanaka factors on engineered poly(ethylene glycol) (PEG) hydrogels. Biomaterials 2013; 34:6559-71. [PMID: 23773820 DOI: 10.1016/j.biomaterials.2013.05.050] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/23/2013] [Indexed: 12/20/2022]
Abstract
Direct reprogramming strategies enable rapid conversion of somatic cells to cardiomyocytes or cardiomyocyte-like cells without going through the pluripotent state. A recently described protocol couples Yamanaka factor induction with pluripotency inhibition followed by BMP4 treatment to achieve rapid reprogramming of mouse fibroblasts to beating cardiomyocyte-like cells. The original study was performed using Matrigel-coated tissue culture polystyrene (TCPS), a stiff material that also non-specifically adsorbs serum proteins. Protein adsorption-resistant poly(ethylene glycol) (PEG) materials can be covalently modified to present precise concentrations of adhesion proteins or peptides without the unintended effects of non-specifically adsorbed proteins. Here, we describe an improved protocol that incorporates custom-engineered materials. We first reproduced the Efe et al. protocol on Matrigel-coated TCPS (the original material), reprogramming adult mouse tail-tip mouse fibroblasts (TTF) and mouse embryonic fibroblasts (MEF) to cardiomyocyte-like cells that demonstrated striated sarcomeric α-actinin staining, spontaneous calcium transients, and visible beating. We then designed poly(ethylene glycol) culture substrates to promote MEF adhesion via laminin and RGD-binding integrins. PEG hydrogels improved proliferation and reprogramming efficiency (evidenced by beating patch number and area, gene expression, and flow cytometry), yielding almost twice the number of sarcomeric α-actinin positive cardiomyocyte-like cells as the originally described substrate. These results illustrate that cellular reprogramming may be enhanced using custom-engineered materials.
Collapse
Affiliation(s)
- Amanda W Smith
- Department of Biomedical Engineering and Center for Materials Innovation, Washington University, Campus Box 1097, One Brookings Dr., St. Louis, MO 63130, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Johnson PJ, Wood MD, Moore AM, Mackinnon SE. Tissue engineered constructs for peripheral nerve surgery. Eur Surg 2013; 45. [PMID: 24385980 DOI: 10.1007/s10353-013-0205-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tissue engineering has been defined as "an interdisciplinary field that applies the principles of engineering and life sciences toward the development of biological substitutes that restore, maintain, or improve tissue function or a whole organ". Traumatic peripheral nerve injury resulting in significant tissue loss at the zone of injury necessitates the need for a bridge or scaffold for regenerating axons from the proximal stump to reach the distal stump. METHODS A review of the literature was used to provide information on the components necessary for the development of a tissue engineered peripheral nerve substitute. Then, a comprehensive review of the literature is presented composed of the studies devoted to this goal. RESULTS Extensive research has been directed toward the development of a tissue engineered peripheral nerve substitute to act as a bridge for regenerating axons from the proximal nerve stump seeking the distal nerve. Ideally this nerve substitute would consist of a scaffold component that mimics the extracellular matrix of the peripheral nerve and a cellular component that serves to stimulate and support regenerating peripheral nerve axons. CONCLUSIONS The field of tissue engineering should consider its challenge to not only meet the autograft "gold standard" but also to understand what drives and inhibits nerve regeneration in order to surpass the results of an autograft.
Collapse
Affiliation(s)
- P J Johnson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, 660 South Euclid, 8238, Saint Louis, MO 63110, USA
| | - M D Wood
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, 660 South Euclid, 8238, Saint Louis, MO 63110, USA
| | - A M Moore
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, 660 South Euclid, 8238, Saint Louis, MO 63110, USA
| | - S E Mackinnon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, 660 South Euclid, 8238, Saint Louis, MO 63110, USA
| |
Collapse
|
47
|
Lu P, Wang Y, Graham L, McHale K, Gao M, Wu D, Brock J, Blesch A, Rosenzweig ES, Havton LA, Zheng B, Conner JM, Marsala M, Tuszynski MH. Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell 2012; 150:1264-73. [PMID: 22980985 DOI: 10.1016/j.cell.2012.08.020] [Citation(s) in RCA: 649] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Revised: 05/02/2012] [Accepted: 08/20/2012] [Indexed: 12/30/2022]
Abstract
Neural stem cells (NSCs) expressing GFP were embedded into fibrin matrices containing growth factor cocktails and grafted to sites of severe spinal cord injury. Grafted cells differentiated into multiple cellular phenotypes, including neurons, which extended large numbers of axons over remarkable distances. Extending axons formed abundant synapses with host cells. Axonal growth was partially dependent on mammalian target of rapamycin (mTOR), but not Nogo signaling. Grafted neurons supported formation of electrophysiological relays across sites of complete spinal transection, resulting in functional recovery. Two human stem cell lines (566RSC and HUES7) embedded in growth-factor-containing fibrin exhibited similar growth, and 566RSC cells supported functional recovery. Thus, properties intrinsic to early-stage neurons can overcome the inhibitory milieu of the injured adult spinal cord to mount remarkable axonal growth, resulting in formation of new relay circuits that significantly improve function. These therapeutic properties extend across stem cell sources and species.
Collapse
Affiliation(s)
- Paul Lu
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Building biocompatible hydrogels for tissue engineering of the brain and spinal cord. J Funct Biomater 2012; 3:839-63. [PMID: 24955749 PMCID: PMC4030922 DOI: 10.3390/jfb3040839] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 10/24/2012] [Indexed: 01/07/2023] Open
Abstract
Tissue engineering strategies employing biomaterials have made great progress in the last few decades. However, the tissues of the brain and spinal cord pose unique challenges due to a separate immune system and their nature as soft tissue. Because of this, neural tissue engineering for the brain and spinal cord may require re-establishing biocompatibility and functionality of biomaterials that have previously been successful for tissue engineering in the body. The goal of this review is to briefly describe the distinctive properties of the central nervous system, specifically the neuroimmune response, and to describe the factors which contribute to building polymer hydrogels compatible with this tissue. These factors include polymer chemistry, polymerization and degradation, and the physical and mechanical properties of the hydrogel. By understanding the necessities in making hydrogels biocompatible with tissue of the brain and spinal cord, tissue engineers can then functionalize these materials for repairing and replacing tissue in the central nervous system.
Collapse
|
49
|
The application of plastic compression to modulate fibrin hydrogel mechanical properties. J Mech Behav Biomed Mater 2012; 16:66-72. [PMID: 23149099 DOI: 10.1016/j.jmbbm.2012.10.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/22/2022]
Abstract
The inherent biocompatibility of fibrin hydrogels makes them an attractive material for use in a wide range of tissue engineering applications. Despite this, their relatively low stiffness and high compliance limits their potential for certain orthopaedic applications. Enhanced mechanical properties are desirable so as to withstand surgical handling and in vivo loading after implantation and additionally, can provide important cues to cells seeded within the hydrogel. Standard methods used to enhance the mechanical properties of biological scaffolds such as chemical or thermal crosslinking cannot be used with fibrin hydrogels as cell seeding and gel formation occurs simultaneously. The objective of this study was to investigate the use of plastic compression as a means to improve the mechanical properties of chondrocyte-seeded fibrin hydrogels and to determine the influence of such compression on cell viability within these constructs. It was found that the application of 80% strain to fibrin hydrogels for 30 min (which resulted in a permanent strain of 47.4%) produced a 2.1-fold increase in the subsequent compressive modulus. Additionally, chondrocyte viability was maintained in the plastically compressed gels with significant cellular proliferation and extracellular matrix accumulation observed over 28 days of culture. In conclusion, plastic compression can be used to modulate the density and mechanical properties of cell-seeded fibrin hydrogels and represents a useful tool for both in theatre and in vitro tissue engineering applications.
Collapse
|
50
|
Oliveira SLB, Pillat MM, Cheffer A, Lameu C, Schwindt TT, Ulrich H. Functions of neurotrophins and growth factors in neurogenesis and brain repair. Cytometry A 2012; 83:76-89. [PMID: 23044513 DOI: 10.1002/cyto.a.22161] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 07/23/2012] [Accepted: 07/31/2012] [Indexed: 12/21/2022]
Abstract
The identification and isolation of multipotent neural stem and progenitor cells in the brain, giving rise to neurons, astrocytes, and oligodendrocytes initiated many studies in order to understand basic mechanisms of endogenous neurogenesis and repair mechanisms of the nervous system and to develop novel therapeutic strategies for cellular regeneration therapies in brain disease. A previous review (Trujillo et al., Cytometry A 2009;75:38-53) focused on the importance of extrinsic factors, especially neurotransmitters, for directing migration and neurogenesis in the developing and adult brain. Here, we extend our review discussing the effects of the principal growth and neurotrophic factors as well as their intracellular signal transduction on neurogenesis, fate determination and neuroprotective mechanisms. Many of these mechanisms have been elucidated by in vitro studies for which neural stem cells were isolated, grown as neurospheres, induced to neural differentiation under desired experimental conditions, and analyzed for embryonic, progenitor, and neural marker expression by flow and imaging cytometry techniques. The better understanding of neural stem cells proliferation and differentiation is crucial for any therapeutic intervention aiming at neural stem cell transplantation and recruitment of endogenous repair mechanisms.
Collapse
Affiliation(s)
- Sophia L B Oliveira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|