1
|
Dill-Macky AS, Lee EN, Wertheim JA, Koss KM. Glia in tissue engineering: From biomaterial tools to transplantation. Acta Biomater 2024; 190:24-49. [PMID: 39396630 DOI: 10.1016/j.actbio.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Glia are imperative in nearly every function of the nervous system, including neurotransmission, neuronal repair, development, immunity, and myelination. Recently, the reparative roles of glia in the central and peripheral nervous systems have been elucidated, suggesting a tremendous potential for these cells as novel treatments to central nervous system disorders. Glial cells often behave as 'double-edged swords' in neuroinflammation, ultimately deciding the life or death of resident cells. Compared to glia, neuronal cells have limited mobility, lack the ability to divide and self-renew, and are generally more delicate. Glia have been candidates for therapeutic use in many successful grafting studies, which have been largely focused on restoring myelin with Schwann cells, olfactory ensheathing glia, and oligodendrocytes with support from astrocytes. However, few therapeutics of this class have succeeded past clinical trials. Several tools and materials are being developed to understand and re-engineer these grafting concepts for greater success, such as extra cellular matrix-based scaffolds, bioactive peptides, biomolecular delivery systems, biomolecular discovery for neuroinflammatory mediation, composite microstructures such as artificial channels for cell trafficking, and graft enhanced electrical stimulation. Furthermore, advances in stem cell-derived cortical/cerebral organoid differentiation protocols have allowed for the generation of patient-derived glia comparable to those acquired from tissues requiring highly invasive procedures or are otherwise inaccessible. However, research on bioengineered tools that manipulate glial cells is nowhere near as comprehensive as that for systems of neurons and neural stem cells. This article explores the therapeutic potential of glia in transplantation with an emphasis on novel bioengineered tools for enhancement of their reparative properties. STATEMENT OF SIGNIFICANCE: Neural glia are responsible for a host of developmental, homeostatic, and reparative roles in the central nervous system but are often a major cause of tissue damage and cellular loss in insults and degenerative pathologies. Most glial grafts have employed Schwann cells for remyelination, but other glial with novel biomaterials have been employed, emphasizing their diverse functionality. Promising strategies have emerged, including neuroimmune mediation of glial scar tissues and facilitated migration and differentiation of stem cells for neural replacement. Herein, a comprehensive review of biomaterial tools for glia in transplantation is presented, highlighting Schwann cells, astrocytes, olfactory ensheating glia, oligodendrocytes, microglia, and ependymal cells.
Collapse
Affiliation(s)
- A S Dill-Macky
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - E N Lee
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - J A Wertheim
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - K M Koss
- Department of Neurobiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0625, United States; Sealy Institute for Drug Discovery, University of Texas Medical Branch, 105 11th Street Galveston, TX 77555-1110, United States.
| |
Collapse
|
2
|
Sicard L, Maillard S, Mbita Akoa D, Torrens C, Collignon AM, Coradin T, Chaussain C. Sclerostin Antibody-Loaded Dense Collagen Hydrogels Promote Critical-Size Bone Defect Repair. ACS Biomater Sci Eng 2024; 10:6451-6464. [PMID: 39269225 DOI: 10.1021/acsbiomaterials.4c00883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
The management of extensive bone loss remains a clinical challenge. Numerous studies are underway to develop a combination of biomaterials, biomolecules, and stem cells to address this challenge. In particular, the systemic administration of antibodies against sclerostin, a regulator of bone formation, was recently shown to enhance the bone repair efficiency of dense collagen hydrogels (DCHs) hosting murine dental pulp stem cells (mDPSCs). The aim of the present study was to assess whether these antibodies, encapsulated and released from DCHs, could promote craniofacial bone repair by the local inhibition of sclerostin. In vitro studies showed that antibody loading modified neither the hydrogel structure nor the viability of seeded mDPSCs. When implanted in a mouse calvaria critical-size bone defect, antibody-loaded DCHs showed repair capabilities similar to those of acellular unloaded DCHs combined with antibody injections. Importantly, the addition of mDPSCs provided no further benefit. Altogether, the local delivery of antisclerostin antibodies from acellular dense collagen scaffolds is highly effective for bone repair. The drastic reduction in the required amount of antibody compared to systemic injection should reduce the cost of the procedure, making the strategy proposed here a promising therapeutic approach for large bone defect repair.
Collapse
Affiliation(s)
- Ludovic Sicard
- Université Paris Cité, Institut des Maladies Musculo-Squelettiques, Orofacial Pathologies, Imaging and Biotherapies Laboratory URP2496 and FHU-DDS-Net, Dental School, and Plateforme d'Imagerie du Vivant (PIV), 92120 Montrouge, France
- AP-HP, Dental Medicine Departments, Bretonneau and Louis Mourier Hospitals, GHN-Université Paris Cité, 75018 Paris, France
| | - Sophie Maillard
- Université Paris Cité, Institut des Maladies Musculo-Squelettiques, Orofacial Pathologies, Imaging and Biotherapies Laboratory URP2496 and FHU-DDS-Net, Dental School, and Plateforme d'Imagerie du Vivant (PIV), 92120 Montrouge, France
| | - Daline Mbita Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), UMR 7574, 4 Place Jussieu, 75005 Paris, France
| | - Coralie Torrens
- Université Paris Cité, Institut des Maladies Musculo-Squelettiques, Orofacial Pathologies, Imaging and Biotherapies Laboratory URP2496 and FHU-DDS-Net, Dental School, and Plateforme d'Imagerie du Vivant (PIV), 92120 Montrouge, France
| | - Anne-Margaux Collignon
- Université Paris Cité, Institut des Maladies Musculo-Squelettiques, Orofacial Pathologies, Imaging and Biotherapies Laboratory URP2496 and FHU-DDS-Net, Dental School, and Plateforme d'Imagerie du Vivant (PIV), 92120 Montrouge, France
- AP-HP, Dental Medicine Departments, Bretonneau and Louis Mourier Hospitals, GHN-Université Paris Cité, 75018 Paris, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris (LCMCP), UMR 7574, 4 Place Jussieu, 75005 Paris, France
| | - Catherine Chaussain
- Université Paris Cité, Institut des Maladies Musculo-Squelettiques, Orofacial Pathologies, Imaging and Biotherapies Laboratory URP2496 and FHU-DDS-Net, Dental School, and Plateforme d'Imagerie du Vivant (PIV), 92120 Montrouge, France
- AP-HP, Dental Medicine Departments, Bretonneau and Louis Mourier Hospitals, GHN-Université Paris Cité, 75018 Paris, France
- AP-HP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, Dental Medicine Department, Bretonneau Hospital, GHN-Université Paris Cité, 75018 Paris, France
| |
Collapse
|
3
|
Yao L, Sai HV, Shippy T, Li B. Cellular and Transcriptional Response of Human Astrocytes to Hybrid Protein Materials. ACS APPLIED BIO MATERIALS 2024; 7:2887-2898. [PMID: 38632900 DOI: 10.1021/acsabm.3c01266] [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] [Indexed: 04/19/2024]
Abstract
Collagen is a major component of the tissue matrix, and soybean can regulate the tissue immune response. Both materials have been used to fabricate biomaterials for tissue repair. In this study, adult and fetal human astrocytes were grown in a soy protein isolate (SPI)-collagen hybrid gel or on the surface of a cross-linked SPI-collagen membrane. Hybrid materials reduced the cell proliferation rate compared to materials generated by collagen alone. However, the hybrid materials did not significantly change the cell motility compared to the control collagen material. RNA-sequencing (RNA-Seq) analysis showed downregulated genes in the cell cycle pathway, including CCNA2, CCNB1, CCNB2, CCND1, CCND2, and CDK1, which may explain lower cell proliferation in the hybrid material. This study also revealed the downregulation of genes encoding extracellular matrix (ECM) components, including HSPG2, LUM, SDC2, COL4A1, COL4A5, COL4A6, and FN1, as well as genes encoding chemokines, including CCL2, CXCL1, CXCL2, CX3CL1, CXCL3, and LIF, for adult human astrocytes grown on the hybrid membrane compared with those grown on the control collagen membrane. The study explored the cellular and transcriptional responses of human astrocytes to the hybrid material and indicated a potential beneficial function of the material in the application of neural repair.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260, United States
| | - Haneesha Vishwa Sai
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260, United States
| | - Teresa Shippy
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, Kansas 66506, United States
| | - Bin Li
- Department of Mechanical Engineering, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260, United States
| |
Collapse
|
4
|
Mbitta Akoa D, Sicard L, Hélary C, Torrens C, Baroukh B, Poliard A, Coradin T. Role of Physico-Chemical and Cellular Conditions on the Bone Repair Potential of Plastically Compressed Collagen Hydrogels. Gels 2024; 10:130. [PMID: 38391460 PMCID: PMC10887598 DOI: 10.3390/gels10020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Since their first description nearly 20 years ago, dense collagen hydrogels obtained by plastic compression have become popular scaffolds in tissue engineering. In particular, when seeded with dental pulp stem cells, they have demonstrated a great in vivo potential in cranial bone repair. Here, we investigated how physico-chemical and cell-seeding conditions could influence the formation and in vitro mineralization of these cellularized scaffolds. A qualitative assessment demonstrated that the gel stability before and after compression was highly sensitive to the conditions of fibrillogenesis, especially initial acid acetic and buffer concentrations. Gels with similar rheological properties but different fibrillar structures that exhibited different stabilities when used for the 3D culture of Stem cells from Human Exfoliated Deciduous teeth (SHEDs) could be prepared. Finally, in our optimal physico-chemical conditions, mineralization could be achieved only using human dental pulp stem cells (hDPSCs) at a high cell density. These results highlight the key role of fibrillogenic conditions and cell type/density on the bone repair potential of cell-laden plastically compressed collagen hydrogels.
Collapse
Affiliation(s)
- Daline Mbitta Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Ludovic Sicard
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
- AP-HP Service de Médecine Bucco-Dentaire, Hôpital Bretonneau, 75018 Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Coralie Torrens
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
| | - Brigitte Baroukh
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
| | - Anne Poliard
- Université de Paris, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, 92120 Montrouge, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| |
Collapse
|
5
|
Hague JP, Andrews AE, Dickinson H. High-throughput design of cultured tissue moulds using a biophysical model: optimising cell alignment. Phys Biol 2023; 20:066006. [PMID: 37899639 DOI: 10.1088/1478-3975/ad0276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/11/2023] [Indexed: 10/31/2023]
Abstract
The technique presented here identifies tethered mould designs, optimised for growing cultured tissue with very highly-aligned cells. It is based on a microscopic biophysical model for polarised cellular hydrogels. There is an unmet need for tools to assist mould and scaffold designs for the growth of cultured tissues with bespoke cell organisations, that can be used in applications such as regenerative medicine, drug screening and cultured meat. High-throughput biophysical calculations were made for a wide variety of computer-generated moulds, with cell-matrix interactions and tissue-scale forces simulated using a contractile network dipole orientation model. Elongated moulds with central broadening and one of the following tethering strategies are found to lead to highly-aligned cells: (1) tethers placed within the bilateral protrusions resulting from an indentation on the short edge, to guide alignment (2) tethers placed within a single vertex to shrink the available space for misalignment. As such, proof-of-concept has been shown for mould and tethered scaffold design based on a recently developed biophysical model. The approach is applicable to a broad range of cell types that align in tissues and is extensible for 3D scaffolds.
Collapse
Affiliation(s)
- James P Hague
- School of Physical Sciences, The Open University, Milton Keynes, MK7 6AA, United Kingdom
| | - Allison E Andrews
- School of Physical Sciences, The Open University, Milton Keynes, MK7 6AA, United Kingdom
| | - Hugh Dickinson
- School of Physical Sciences, The Open University, Milton Keynes, MK7 6AA, United Kingdom
| |
Collapse
|
6
|
Achenbach P, Hillerbrand L, Gerardo-Nava JL, Dievernich A, Hodde D, Sechi AS, Dalton PD, Pich A, Weis J, Altinova H, Brook GA. Function Follows Form: Oriented Substrate Nanotopography Overrides Neurite-Repulsive Schwann Cell-Astrocyte Barrier Formation in an In Vitro Model of Glial Scarring. NANO LETTERS 2023; 23:6337-6346. [PMID: 37459449 DOI: 10.1021/acs.nanolett.3c00873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Schwann cell (SC) transplantation represents a promising therapeutic approach for traumatic spinal cord injury but is frustrated by barrier formation, preventing cell migration, and axonal regeneration at the interface between grafted SCs and reactive resident astrocytes (ACs). Although regenerating axons successfully extend into SC grafts, only a few cross the SC-AC interface to re-enter lesioned neuropil. To date, research has focused on identifying and modifying the molecular mechanisms underlying such scarring cell-cell interactions, while the influence of substrate topography remains largely unexplored. Using a recently modified cell confrontation assay to model SC-AC barrier formation in vitro, highly oriented poly(ε-caprolactone) nanofibers were observed to reduce AC reactivity, induce extensive oriented intermingling between SCs and ACs, and ultimately enable substantial neurite outgrowth from the SC compartment into the AC territory. It is anticipated that these findings will have important implications for the future design of biomaterial-based scaffolds for nervous tissue repair.
Collapse
Affiliation(s)
- Pascal Achenbach
- Department of Neurology, RWTH Aachen University Hospital, 52074 Aachen, Germany
- Institute of Neuropathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Laura Hillerbrand
- Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, 97070 Würzburg, Germany
| | - José L Gerardo-Nava
- DWI - Leibniz Institute for Interactive Materials, 52074 Aachen, Germany
- Advanced Materials for Biomedicine (AMB), Institute of Applied Medical Engineering (AME), RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Axel Dievernich
- FEG Textiltechnik Forschungs- und Entwicklungsgesellschaft mbH, 52070 Aachen, Germany
| | - Dorothee Hodde
- Institute of Neuropathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
- University Hospital, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | - Antonio S Sechi
- Department of Cell and Tumor Biology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Paul D Dalton
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon 97403, United States
| | - Andrij Pich
- DWI - Leibniz Institute for Interactive Materials, 52074 Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Haktan Altinova
- Institute of Neuropathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
- Department of Neurosurgery, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Gary A Brook
- Institute of Neuropathology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
7
|
Mogas Barcons A, Chowdhury F, Chari DM, Adams C. Systematic Alignment Analysis of Neural Transplant Cells in Electrospun Nanofibre Scaffolds. MATERIALS (BASEL, SWITZERLAND) 2022; 16:124. [PMID: 36614463 PMCID: PMC9821626 DOI: 10.3390/ma16010124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Spinal cord injury is debilitating with functional loss often permanent due to a lack of neuro-regenerative or neuro-therapeutic strategies. A promising approach to enhance biological function is through implantation of tissue engineered constructs, to offer neural cell replacement and reconstruction of the functional neuro-architecture. A key goal is to achieve spatially targeted guidance of regenerating tissue across the lesion site to achieve an aligned tissue structure lost as a consequence of injury. Electrospun nanofibres mimic the nanoscale architecture of the spinal cord, can be readily aligned, functionalised with pro-regenerative molecules and incorporated into implantable matrices to provide topographical cues. Crucially, electrospun nanofibers are routinely manufactured at a scale required for clinical use. Although promising, few studies have tested whether electrospun nanofibres can guide targeted spatial growth of clinically relevant neural stem/precursor populations. The alignment fate of daughter cells (derived from the pre-aligned parent cells) has also received limited attention. Further, a standardised quantification methodology to correlate neural cell alignment with topographical cues is not available. We have adapted an image analysis technique to quantify nanofibre-induced alignment of neural cells. Using this method, we show that two key neural stem/precursor populations of clinical relevance (namely, neural stem cells (NSCs) and oligodendrocyte precursor cells), reproducibly orientate their growth to aligned, high-density electrospun nanofiber meshes, but not randomly distributed ones. Daughter populations derived from aligned NSCs (neurons and astrocytes) maintained their alignment following differentiation, but oligodendrocytes did not. Our data show that pre-aligned transplant populations can be used to generate complex, multicellular aligned-fibre constructs for neural implantation.
Collapse
Affiliation(s)
- Aina Mogas Barcons
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | | | - Divya M. Chari
- School of Medicine, Keele University, Newcastle-under-Lyme ST5 5BG, UK
| | - Christopher Adams
- School of Life Sciences, Keele University, Newcastle-under-Lyme ST5 5BG, UK
| |
Collapse
|
8
|
Rayner MLD, Kellaway SC, Kingston I, Guillemot-Legris O, Gregory H, Healy J, Phillips JB. Exploring the Nerve Regenerative Capacity of Compounds with Differing Affinity for PPARγ In Vitro and In Vivo. Cells 2022; 12:cells12010042. [PMID: 36611836 PMCID: PMC9818498 DOI: 10.3390/cells12010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Damage to peripheral nerves can cause debilitating consequences for patients such as lifelong pain and disability. At present, no drug treatments are routinely given in the clinic following a peripheral nerve injury (PNI) to improve regeneration and remyelination of damaged nerves. Appropriately targeted therapeutic agents have the potential to be used at different stages following nerve damage, e.g., to maintain Schwann cell viability, induce and sustain a repair phenotype to support axonal growth, or promote remyelination. The development of therapies to promote nerve regeneration is currently of high interest to researchers, however, translation to the clinic of drug therapies for PNI is still lacking. Studying the effect of PPARγ agonists for treatment of peripheral nerve injures has demonstrated significant benefits. Ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), has reproducibly demonstrated benefits in vitro and in vivo, suggested to be due to its agonist action on PPARγ. Other NSAIDs have demonstrated differing levels of PPARγ activation based upon their affinity. Therefore, it was of interest to determine whether affinity for PPARγ of selected drugs corresponded to an increase in regeneration. A 3D co-culture in vitro model identified some correlation between these two properties. However, when the drug treatments were screened in vivo, in a crush injury model in a rat sciatic nerve, the same correlation was not apparent. Further differences were observed between capacity to increase axon number and improvement in functional recovery. Despite there not being a clear correlation between affinity and size of effect on regeneration, all selected PPARγ agonists improved regeneration, providing a panel of compounds that could be explored for use in the treatment of PNI.
Collapse
Affiliation(s)
- Melissa L. D. Rayner
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
- Correspondence:
| | - Simon C. Kellaway
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Isabel Kingston
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Owein Guillemot-Legris
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Holly Gregory
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Jess Healy
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| |
Collapse
|
9
|
Roberton VH, Phillips JB. Considerations for the use of biomaterials to support cell therapy in neurodegenerative disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:191-205. [DOI: 10.1016/bs.irn.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
10
|
Mezzasalma SA, Grassi L, Grassi M. Physical and chemical properties of carbon nanotubes in view of mechanistic neuroscience investigations. Some outlook from condensed matter, materials science and physical chemistry. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112480. [PMID: 34857266 DOI: 10.1016/j.msec.2021.112480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/08/2021] [Accepted: 10/07/2021] [Indexed: 01/17/2023]
Abstract
The open border between non-living and living matter, suggested by increasingly emerging fields of nanoscience interfaced to biological systems, requires a detailed knowledge of nanomaterials properties. An account of the wide spectrum of phenomena, belonging to physical chemistry of interfaces, materials science, solid state physics at the nanoscale and bioelectrochemistry, thus is acquainted for a comprehensive application of carbon nanotubes interphased with neuron cells. This review points out a number of conceptual tools to further address the ongoing advances in coupling neuronal networks with (carbon) nanotube meshworks, and to deepen the basic issues that govern a biological cell or tissue interacting with a nanomaterial. Emphasis is given here to the properties and roles of carbon nanotube systems at relevant spatiotemporal scales of individual molecules, junctions and molecular layers, as well as to the point of view of a condensed matter or materials scientist. Carbon nanotube interactions with blood-brain barrier, drug delivery, biocompatibility and functionalization issues are also regarded.
Collapse
Affiliation(s)
- Stefano A Mezzasalma
- Ruder Bošković Institute, Materials Physics Division, Bijeniška cesta 54, 10000 Zagreb, Croatia; Lund Institute for advanced Neutron and X-ray Science (LINXS), Lund University, IDEON Building, Delta 5, Scheelevägen 19, 223 70 Lund, Sweden.
| | - Lucia Grassi
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, via Valerio 6, I-34127 Trieste, Italy.
| |
Collapse
|
11
|
Capturing the third dimension in drug discovery: Spatially-resolved tools for interrogation of complex 3D cell models. Biotechnol Adv 2021; 55:107883. [PMID: 34875362 DOI: 10.1016/j.biotechadv.2021.107883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
Advanced three-dimensional (3D) cell models have proven to be capable of depicting architectural and microenvironmental features of several tissues. By providing data of higher physiological and pathophysiological relevance, 3D cell models have been contributing to a better understanding of human development, pathology onset and progression mechanisms, as well as for 3D cell-based assays for drug discovery. Nonetheless, the characterization and interrogation of these tissue-like structures pose major challenges on the conventional analytical methods, pushing the development of spatially-resolved technologies. Herein, we review recent advances and pioneering technologies suitable for the interrogation of multicellular 3D models, while capable of retaining biological spatial information. We focused on imaging technologies and omics tools, namely transcriptomics, proteomics and metabolomics. The advantages and shortcomings of these novel methodologies are discussed, alongside the opportunities to intertwine data from the different tools.
Collapse
|
12
|
Nicaise AM, D'Angelo A, Ionescu RB, Krzak G, Willis CM, Pluchino S. The role of neural stem cells in regulating glial scar formation and repair. Cell Tissue Res 2021; 387:399-414. [PMID: 34820704 PMCID: PMC8975756 DOI: 10.1007/s00441-021-03554-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Glial scars are a common pathological occurrence in a variety of central nervous system (CNS) diseases and injuries. They are caused after severe damage and consist of reactive glia that form a barrier around the damaged tissue that leads to a non-permissive microenvironment which prevents proper endogenous regeneration. While there are a number of therapies that are able to address some components of disease, there are none that provide regenerative properties. Within the past decade, neural stem cells (NSCs) have been heavily studied due to their potent anti-inflammatory and reparative capabilities in disease and injury. Exogenously applied NSCs have been found to aid in glial scar healing by reducing inflammation and providing cell replacement. However, endogenous NSCs have also been found to contribute to the reactive environment by different means. Further understanding how NSCs can be leveraged to aid in the resolution of the glial scar is imperative in the use of these cells as regenerative therapies. To do so, humanised 3D model systems have been developed to study the development and maintenance of the glial scar. Herein, we explore the current work on endogenous and exogenous NSCs in the glial scar as well as the novel 3D stem cell–based technologies being used to model this pathology in a dish.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK.
| | - Andrea D'Angelo
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Rosana-Bristena Ionescu
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Grzegorz Krzak
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Cory M Willis
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Abraham JA, Blaschke S, Tarazi S, Dreissen G, Vay SU, Schroeter M, Fink GR, Merkel R, Rueger MA, Hoffmann B. NSCs Under Strain-Unraveling the Mechanoprotective Role of Differentiating Astrocytes in a Cyclically Stretched Coculture With Differentiating Neurons. Front Cell Neurosci 2021; 15:706585. [PMID: 34630042 PMCID: PMC8497758 DOI: 10.3389/fncel.2021.706585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/31/2021] [Indexed: 11/19/2022] Open
Abstract
The neural stem cell (NSC) niche is a highly vascularized microenvironment that supplies stem cells with relevant biological and chemical cues. However, the NSCs’ proximity to the vasculature also means that the NSCs are subjected to permanent tissue deformation effected by the vessels’ heartbeat-induced pulsatile movements. Cultivating NSCs under common culture conditions neglects the—yet unknown—influence of this cyclic mechanical strain on neural stem cells. Under the hypothesis that pulsatile strain should affect essential NSC functions, a cyclic uniaxial strain was applied under biomimetic conditions using an in-house developed stretching system based on cross-linked polydimethylsiloxane (PDMS) elastomer. While lineage commitment remained unaffected by cyclic deformation, strain affected NSC quiescence and cytoskeletal organization. Unexpectedly, cyclically stretched stem cells aligned in stretch direction, a phenomenon unknown for other types of cells in the mammalian organism. The same effect was observed for young astrocytes differentiating from NSCs. In contrast, young neurons differentiating from NSCs did not show mechanoresponsiveness. The exceptional orientation of NSCs and young astrocytes in the stretch direction was blocked upon RhoA activation and went along with a lack of stress fibers. Compared to postnatal astrocytes and mature neurons, NSCs and their young progeny displayed characteristic and distinct mechanoresponsiveness. Data suggest a protective role of young astrocytes in mixed cultures of differentiating neurons and astrocytes by mitigating the mechanical stress of pulsatile strain on developing neurons.
Collapse
Affiliation(s)
- Jella-Andrea Abraham
- Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Stefan Blaschke
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Samar Tarazi
- Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Georg Dreissen
- Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Sabine U Vay
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany
| | - Michael Schroeter
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Rudolf Merkel
- Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Maria A Rueger
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Bernd Hoffmann
- Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| |
Collapse
|
14
|
In vitro model of traumatic brain injury to screen neuro-regenerative biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112253. [PMID: 34474815 DOI: 10.1016/j.msec.2021.112253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/21/2021] [Accepted: 06/11/2021] [Indexed: 11/24/2022]
Abstract
Penetrating traumatic brain injury (pTBI) causes serious neurological deficits with no clinical regenerative therapies currently available. Tissue engineering strategies using biomaterial-based 'structural bridges' offer high potential to promote neural regeneration post-injury. This includes surgical grade materials which can be repurposed as biological scaffolds to overcome challenges associated with long approval processes and scaleup for human application. However, high throughput, pathomimetic models of pTBI are lacking for the developmental testing of such neuro-materials, representing a bottleneck in this rapidly emergent field. We have established a high throughput and facile culture model containing the major neural cell types which govern biomaterial handling in the central nervous system. We show that induction of traumatic injuries was feasible in the model, with post-injury implantation of a surgical grade biomaterial. Cellular imaging in lesions was achievable using standard epifluorescence microscopy methods. Key pathological features of pTBI were evident in vitro namely immune cell infiltration of lesions/biomaterial, with responses characteristic of cell scarring, namely hypertrophic astrocytes with GFAP upregulation. Based on our observations, we consider the high-throughput, inexpensive and facile pTBI model can be used to study biomaterial 'implantation' and evaluate neural cell-biomaterial responses. The model is highly versatile to test a range of laboratory and clinical grade materials for neural regeneration.
Collapse
|
15
|
Yaqub N, Wayne G, Birchall M, Song W. Recent advances in human respiratory epithelium models for drug discovery. Biotechnol Adv 2021; 54:107832. [PMID: 34481894 DOI: 10.1016/j.biotechadv.2021.107832] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/08/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
The respiratory epithelium is intimately associated with the pathophysiologies of highly infectious viral contagions and chronic illnesses such as chronic obstructive pulmonary disorder, presently the third leading cause of death worldwide with a projected economic burden of £1.7 trillion by 2030. Preclinical studies of respiratory physiology have almost exclusively utilised non-humanised animal models, alongside reductionistic cell line-based models, and primary epithelial cell models cultured at an air-liquid interface (ALI). Despite their utility, these model systems have been limited by their poor correlation to the human condition. This has undermined the ability to identify novel therapeutics, evidenced by a 15% chance of success for medicinal respiratory compounds entering clinical trials in 2018. Consequently, preclinical studies require new translational efficacy models to address the problem of respiratory drug attrition. This review describes the utility of the current in vivo (rodent), ex vivo (isolated perfused lungs and precision cut lung slices), two-dimensional in vitro cell-line (A549, BEAS-2B, Calu-3) and three-dimensional in vitro ALI (gold-standard and co-culture) and organoid respiratory epithelium models. The limitations to the application of these model systems in drug discovery research are discussed, in addition to perspectives of the future innovations required to facilitate the next generation of human-relevant respiratory models.
Collapse
Affiliation(s)
- Naheem Yaqub
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK
| | - Gareth Wayne
- Novel Human Genetics, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Martin Birchall
- The Ear Institute, Faculty of Brain Sciences, University College London, London WC1X 8EE, UK.
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK.
| |
Collapse
|
16
|
Gordián-Vélez WJ, Chouhan D, España RA, Chen HI, Burdick JA, Duda JE, Cullen DK. Restoring lost nigrostriatal fibers in Parkinson's disease based on clinically-inspired design criteria. Brain Res Bull 2021; 175:168-185. [PMID: 34332016 DOI: 10.1016/j.brainresbull.2021.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022]
Abstract
Parkinson's disease is a neurodegenerative disease affecting around 10 million people worldwide. The death of dopaminergic neurons in the substantia nigra and the axonal fibers that constitute the nigrostriatal pathway leads to a loss of dopamine in the striatum that causes the motor symptoms of this disease. Traditional treatments have focused on reducing symptoms, while therapies with human fetal or stem cell-derived neurons have centered on implanting these cells in the striatum to restore its innervation. An alternative approach is pathway reconstruction, which aims to rebuild the entire structure of neurons and axonal fibers of the nigrostriatal pathway in a way that matches its anatomy and physiology. This type of repair could be more capable of reestablishing the signaling mechanisms that ensure proper dopamine release in the striatum and regulation of other motor circuit regions in the brain. In this manuscript, we conduct a review of the literature related to pathway reconstruction as a treatment for Parkinson's disease, delve into the limitations of these studies, and propose the requisite design criteria to achieve this goal at a human scale. We then present our tissue engineering-based platform to fabricate hydrogel-encased dopaminergic axon tracts in vitro for later implantation into the brain to replace and reconstruct the pathway. These tissue-engineered nigrostriatal pathways (TE-NSPs) can be characterized and optimized for cell number and phenotype, axon growth lengths and rates, and the capacity for synaptic connectivity and dopamine release. We then show original data of advances in creating these constructs matching clinical design criteria using human iPSC-derived dopaminergic neurons and a hyaluronic acid hydrogel. We conclude with a discussion of future steps that are needed to further optimize human-scale TE-NSPs and translate them into clinical products.
Collapse
Affiliation(s)
- Wisberty J Gordián-Vélez
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Dimple Chouhan
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Rodrigo A España
- Department of Neurobiology & Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - H Isaac Chen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - John E Duda
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D Kacy Cullen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States.
| |
Collapse
|
17
|
Keskin-Erdogan Z, Patel KD, Chau DYS, Day RM, Kim HW, Knowles JC. Utilization of GelMA with phosphate glass fibers for glial cell alignment. J Biomed Mater Res A 2021; 109:2212-2224. [PMID: 33960663 DOI: 10.1002/jbm.a.37206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/15/2022]
Abstract
Glial cell alignment in tissue engineered constructs is essential for achieving functional outcomes in neural recovery. While gelatin methacrylate (GelMA) hydrogel offers superior biocompatibility along with permissive structure and tailorable mechanical properties, phosphate glass fibers (PGFs) can provide physical cues for directionality of neural growth. Aligned PGFs were fabricated by a melt quenching and fiber drawing method and utilized with synthesized GelMA hydrogel. The mechanical properties of GelMA and biocompatibility of the GelMA-PGFs composite were investigated in vitro using rat glial cells. GelMA with 86% methacrylation degree were photo-crosslinked using 0.1%wt photo-initiator (PI). Photocrosslinking under UV exposure for 60 s was used to produce hydrogels (GelMA-60). PGFs were introduced into the GelMA before crosslinking. Storage modulus and loss modulus of GelMA-60 was 24.73 ± 2.52 and 1.08 ± 0.23 kN/m2 , respectively. Increased cell alignment was observed in GelMA-PGFs compared with GelMA hydrogel alone. These findings suggest GelMA-PGFs can provide glial cells with physical cues necessary to achieve cell alignment. This approach could further be used to achieve glial cell alignment in bioengineered constructs designed to bridge damaged nerve tissue.
Collapse
Affiliation(s)
- Zalike Keskin-Erdogan
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, London, UK
| | - Kapil D Patel
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea
| | - David Y S Chau
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, London, UK.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea
| | - Richard M Day
- Centre for Precision Healthcare, UCL Division of Medicine, University College London, London, UK
| | - Hae-Won Kim
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Republic of Korea
| | - Jonathan C Knowles
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, London, UK.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
18
|
Structural and functional properties of astrocytes on PCL based electrospun fibres. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111363. [DOI: 10.1016/j.msec.2020.111363] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/03/2020] [Accepted: 08/03/2020] [Indexed: 01/18/2023]
|
19
|
Morphological Changes in Astrocytes by Self-Oxidation of Dopamine to Polydopamine and Quantification of Dopamine through Multivariate Regression Analysis of Polydopamine Images. Polymers (Basel) 2020; 12:polym12112483. [PMID: 33114705 PMCID: PMC7692622 DOI: 10.3390/polym12112483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/10/2023] Open
Abstract
Astrocytes, also known as astroglia, are important cells for the structural support of neurons as well as for biochemical balance in the central nervous system (CNS). In this study, the polymerization of dopamine (DA) to polydopamine (PDA) and its effect on astrocytes was investigated. The polymerization of DA, being directly proportional to the DA concentration, raises the prospect of detecting DA concentration from PDA optically using image-processing techniques. It was found here that DA, a naturally occurring neurotransmitter, significantly altered astrocyte cell number, morphology, and metabolism, compared to astrocytes in the absence of DA. Along with these effects on astrocytes, the polymerization of DA to PDA was tracked optically in the same cell culture wells. This polymerization process led to a unique methodology based on multivariate regression analysis that quantified the concentration of DA from optical images of astrocyte cell culture media. Therefore, this developed methodology, combined with conventional imaging equipment, could be used in place of high-end and expensive analytical chemistry instruments, such as spectrophotometry, mass spectrometry, and fluorescence techniques, for quantification of the concentration of DA after polymerization to PDA under in vitro and potentially in vivo conditions.
Collapse
|
20
|
Kim BJ, Choi JY, Choi H, Han S, Seo J, Kim J, Joo S, Kim HM, Oh C, Hong S, Kim P, Choi IS. Astrocyte-Encapsulated Hydrogel Microfibers Enhance Neuronal Circuit Generation. Adv Healthc Mater 2020; 9:e1901072. [PMID: 31957248 DOI: 10.1002/adhm.201901072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/23/2019] [Indexed: 12/18/2022]
Abstract
Astrocytes, the most representative glial cells in the brain, play a multitude of crucial functions for proper neuronal development and synaptic-network formation, including neuroprotection as well as physical and chemical support. However, little attention has been paid, in the neuroregenerative medicine and related fields, to the cytoprotective incorporation of astrocytes into neuron-culture scaffolds and full-fledged functional utilization of encapsulated astrocytes for controlled neuronal development. In this article, a 3D neurosupportive culture system for enhanced induction of neuronal circuit generation is reported, where astrocytes are confined in hydrogel microfibers and protected from the outside. The astrocyte-encapsulated microfibers significantly accelerate the neurite outgrowth and guide its directionality, and enhance the synaptic formation, without any physical contact with the neurons. This astrocyte-laden system provides a pivotal culture scaffold for advanced development of cell-based therapeutics for neural injuries, such as spinal cord injury.
Collapse
Affiliation(s)
- Beom Jin Kim
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Ji Yu Choi
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Hyunwoo Choi
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Sol Han
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Jeongyeon Seo
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Jungnam Kim
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Sunghoon Joo
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| | - Hyo Min Kim
- Department of Bio and Brain EngineeringKAIST Daejeon 34141 Korea
| | - Chungik Oh
- Department of Materials Science and EngineeringKAIST Daejeon 34141 Korea
| | - Seungbum Hong
- Department of Materials Science and EngineeringKAIST Daejeon 34141 Korea
| | - Pilnam Kim
- Department of Bio and Brain EngineeringKAIST Daejeon 34141 Korea
| | - Insung S. Choi
- Center for Cell‐Encapsulation ResearchDepartment of ChemistryKAIST Daejeon 34141 Korea
| |
Collapse
|
21
|
Decellularized brain matrix enhances macrophage polarization and functional improvements in rat spinal cord injury. Acta Biomater 2020; 101:357-371. [PMID: 31711898 DOI: 10.1016/j.actbio.2019.11.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/03/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is a devastating lesion lacking effective treatment options currently available in clinics. The inflammatory process exacerbates the extent of the lesion through a secondary injury mechanism, where proinflammatory classically activated macrophages (M1) are prevalent at the lesion site. However, the polarized alternatively activated anti-inflammatory macrophages (M2) are known to play an important role in wound healing and regeneration following SCI. Herein, we introduce porcine brain decellularized extracellular matrix (dECM) to modulate the macrophages in the injured spinal cord. The hydrogels with collagen and dECM at various dECM concentrations (1, 5, and 8 mg/ml) were used to cultivate primary macrophages and neurons. The dECM hydrogels were shown to promote the polarization of macrophages toward M2 phase and the neurite outgrowth of cortical and hippocampal neurons. When the dECM hydrogels were applied to rat SCI models, the proportion of M1 and M2 macrophages in the injured spinal cord was substantially altered. When received dECM concetration of 5 mg/ml, the expression of molecules associated with M2 (CD206, arginase1, and IL-10) was significantly increased. Consistently, the population of total macrophages and cavity area were substantially reduced in the dECM-treated groups. As a result, the locomotor functions of injured spinal cord, as assessed by BBB and ladder scoring, were significantly improved. Collectively, the porcine brain dECM with optimal concentration promotes functional recovery in SCI models through the activation of M2 macrophages, suggesting the promising use of the engineered hydrogels in the treatment of acute SCI. STATEMENT OF SIGNIFICANCE: Spinal cord injury (SCI) is a devastating lesion, lacking effective treatment options currently available in clinics. Here we delineated that the treatment of injured spinal cord with porcine brain decellularized matrix-based hydrogels for the first time, and could modulate the macrophage polarization and the ultimate functional recovery. When appropriate formulations were applied to a contused spinal cord model in rats, the decellularized matrix hydrogels shifted the macrophages to polarize to pro-regenerative M2 phenotype, decreased the size of lesion cavity, and finally promoted the locomotor functions until 8 weeks following the injury. We consider this work can significantly augment the matrix(biomaterial)-based therapeutic options, as an alternative to drug or cell-free approaches, for the treatment of acute injury of spinal cord.
Collapse
|
22
|
Johnson CDL, Zuidema JM, Kearns KR, Maguire AB, Desmond GP, Thompson DM, Gilbert RJ. The Effect of Electrospun Fiber Diameter on Astrocyte-Mediated Neurite Guidance and Protection. ACS APPLIED BIO MATERIALS 2018; 2:104-117. [PMID: 31061987 DOI: 10.1021/acsabm.8b00432] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The topography of electrospun fiber scaffolds modifies astrocytes toward in vivo-like morphologies and behaviors. However, little is known about how electrospun fiber diameter influences astrocyte behavior. In this work, aligned fibers with two distinct nanoscale fiber diameters (808 and 386 nm) were prepared, and the astrocyte response was measured over time. Astrocytes on the large diameter fibers showed significantly increased elongation as early as 2 h after seeding and remained significantly more elongated for up to 4 days compared to those on small diameter fibers. Astrocytes extending along larger diameter fibers were better equipped to support long neurite outgrowth from dorsal root ganglia neurons, and neurite outgrowth along these astrocytes was less branched than outgrowth along astrocytes cultured on small diameter fibers. The differences in astrocyte shape observed on the small or large diameter fibers did not translate into differences in GLT-1, GFAP, or GLAST protein expression. Thus, different fiber diameters were unable to influence astrocyte protein expression uniquely. Nevertheless, astrocytes cultured in either small or large fibers significantly increased their expression of GLT-1 compared to astrocytes cultured on nonfiber (film) controls. Fibrous-induced increases in astrocyte GLT-1 expression protected astrocyte/neuron cocultures from toxicity generated by high extracellular glutamate. Alternatively, astrocytes/neurons cultured on films were less able to protect these cells from culture conditions consisting of high glutamate levels. Biomaterials, such as the fibrous materials presented here, may help stimulate astrocytes to increase GLT-1 expression and uptake more glutamate, since astrocytes are less likely to uptake glutamate in neurodegenerative pathologies or following central nervous system injury.
Collapse
Affiliation(s)
- Christopher D L Johnson
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States
| | - Jonathan M Zuidema
- Department of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
| | - Kathryn R Kearns
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States
| | - Alianna B Maguire
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States
| | - Gregory P Desmond
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States
| | - Deanna M Thompson
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States
| | - Ryan J Gilbert
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 Eighth Street, Troy, New York 12180-3590, United States
| |
Collapse
|
23
|
Rayner MLD, Laranjeira S, Evans RE, Shipley RJ, Healy J, Phillips JB. Developing an In Vitro Model to Screen Drugs for Nerve Regeneration. Anat Rec (Hoboken) 2018; 301:1628-1637. [PMID: 30334365 PMCID: PMC6282521 DOI: 10.1002/ar.23918] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/24/2018] [Accepted: 03/25/2018] [Indexed: 12/29/2022]
Abstract
Peripheral nerve injuries (PNI) have a high prevalence and can be debilitating, resulting in life‐long loss or disturbance in end‐organ function, which compromises quality of life for patients. Current therapies use microsurgical approaches but there is the potential for enhancing recovery through other therapeutic modalities such as; cell‐based conduits, gene therapy and small molecules. A number of molecular targets and drugs which have the potential to improve nerve regeneration have been identified, however, there are challenges associated with moving therapies toward clinical translation. Due to the lack of detailed knowledge about the pro‐regenerative effect of potential drug treatments, there is a need for effective in vitro models to screen compounds to inform future pre‐clinical and clinical studies. The interaction between regenerating neurites and supporting Schwann cells is a key feature of the nerve environment, therefore, in vitro models that mimic this cellular association are useful tools. In this study, we have investigated various cell culture models, including simple monolayer systems and more complex 3D‐engineered co‐cultures, as models for use in PNI drug development. Anat Rec, 301:1628–1637, 2018. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Melissa L D Rayner
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, WC1X 8LD, London, UK.,Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| | - Simão Laranjeira
- UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK.,UCL Department of Mechanical Engineering, University College London, WC1E 7JEK, London, UK
| | - Rachael E Evans
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, WC1X 8LD, London, UK.,Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| | - Rebecca J Shipley
- UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK.,UCL Department of Mechanical Engineering, University College London, WC1E 7JEK, London, UK
| | - Jess Healy
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| | - James B Phillips
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, WC1X 8LD, London, UK.,Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| |
Collapse
|
24
|
Lichtenstein MP, Carretero NM, Pérez E, Pulido-Salgado M, Moral-Vico J, Solà C, Casañ-Pastor N, Suñol C. Biosafety assessment of conducting nanostructured materials by using co-cultures of neurons and astrocytes. Neurotoxicology 2018; 68:115-125. [PMID: 30031109 DOI: 10.1016/j.neuro.2018.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 07/05/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022]
Abstract
Neural electrode implants are made mostly of noble materials. We have synthesized a nanostructured material combining the good electrochemical properties of iridium oxide (IrOx) and carbon-nanotubes (CNT) and the properties of poly(3,4-ethylenedioxythiophene) (PEDOT). IrOx-CNT-PEDOT charge storage capacity was lower than that of IrOx and IrOx-CNT, but higher than that of other PEDOT-containing hybrids and Pt. Cyclic voltammetry, SEM, XPS and micro-Raman spectroscopy suggest that PEDOT encapsulates IrOx and CNT. In our search for a cell culture platform that could optimize modelling the in vivo environment, we determined cell viability, neuron and astrocyte functionality and the response of astrocytes to an inflammatory insult by using primary cultures of neurons, of astrocytes and co-cultures of both. The materials tested (based on IrOx, CNT and PEDOT, as well as Pt as a reference) allowed adhesion and proliferation of astrocytes and full compatibility for neurons grown in co-cultures. Functionality assays show that uptake of glutamate in neuron-astrocyte co-culture was significantly higher than the sum of the uptake in astrocytes and neurons. In co-cultures on IrOx, IrOx-CNT and IrOx-CNT-PEDOT, glutamate was released by a depolarizing stimulus and induced a significant increase in intracellular calcium, supporting the expression of functional NMDA/glutamate receptors. LPS-induced inflammatory response in astrocytes showed a decreased response in NOS2 and COX2 mRNA expression for IrOx-CNT-PEDOT. Results indicate that neuron-astrocyte co-cultures are a reliable model for assessing the biocompatibility and safety of nanostructured materials, evidencing also that hybrid IrOx-CNT-PEDOT nanocomposite materials may offer larger resistance to inflammatory insults.
Collapse
Affiliation(s)
- Mathieu P Lichtenstein
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB, CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c/Rosselló 161, 08036 Barcelona, Spain
| | - Nina M Carretero
- Institut de Ciències de Materials de Barcelona (ICMAB, CSIC), Campus UAB, E-08193 Bellaterra, Barcelona, Spain
| | - Estela Pérez
- Institut de Ciències de Materials de Barcelona (ICMAB, CSIC), Campus UAB, E-08193 Bellaterra, Barcelona, Spain
| | - Marta Pulido-Salgado
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB, CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c/Rosselló 161, 08036 Barcelona, Spain
| | - Javier Moral-Vico
- Institut de Ciències de Materials de Barcelona (ICMAB, CSIC), Campus UAB, E-08193 Bellaterra, Barcelona, Spain
| | - Carme Solà
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB, CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c/Rosselló 161, 08036 Barcelona, Spain
| | - Nieves Casañ-Pastor
- Institut de Ciències de Materials de Barcelona (ICMAB, CSIC), Campus UAB, E-08193 Bellaterra, Barcelona, Spain.
| | - Cristina Suñol
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB, CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c/Rosselló 161, 08036 Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Spain.
| |
Collapse
|
25
|
Tickle JA, Poptani H, Taylor A, Chari DM. Noninvasive imaging of nanoparticle-labeled transplant populations within polymer matrices for neural cell therapy. Nanomedicine (Lond) 2018; 13:1333-1348. [PMID: 29949467 PMCID: PMC6220152 DOI: 10.2217/nnm-2017-0347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/29/2018] [Indexed: 12/15/2022] Open
Abstract
AIM To develop a 3D neural cell construct for encapsulated delivery of transplant cells; develop hydrogels seeded with magnetic nanoparticle (MNP)-labeled cells suitable for cell tracking by MRI. MATERIALS & METHODS Astrocytes were exogenously labeled with MRI-compatible iron-oxide MNPs prior to intra-construct incorporation within a 3D collagen hydrogel. RESULTS A connective, complex cellular network was clearly observable within the 3D constructs, with high cellular viability. MNP accumulation in astrocytes provided a hypointense MRI signal at 24 h & 14 days. CONCLUSION Our findings support the concept of developing a 3D construct possessing the dual advantages of (i) support of long-term cell survival of neural populations with (ii) the potential for noninvasive MRI-tracking of intra-construct cells for neuroregenerative applications.
Collapse
Affiliation(s)
- Jacqueline A Tickle
- Institute for Science & Technology in Medicine, Keele University, Keele, ST5 5BG, UK
| | - Harish Poptani
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Arthur Taylor
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Divya M Chari
- Institute for Science & Technology in Medicine, Keele University, Keele, ST5 5BG, UK
| |
Collapse
|
26
|
Chong SA, Balosso S, Vandenplas C, Szczesny G, Hanon E, Claes K, Van Damme X, Danis B, Van Eyll J, Wolff C, Vezzani A, Kaminski RM, Niespodziany I. Intrinsic Inflammation Is a Potential Anti-Epileptogenic Target in the Organotypic Hippocampal Slice Model. Neurotherapeutics 2018; 15:470-488. [PMID: 29464573 PMCID: PMC5935638 DOI: 10.1007/s13311-018-0607-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Understanding the mechanisms of epileptogenesis is essential to develop novel drugs that could prevent or modify the disease. Neuroinflammation has been proposed as a promising target for therapeutic interventions to inhibit the epileptogenic process that evolves from traumatic brain injury. However, it remains unclear whether cytokine-related pathways, particularly TNFα signaling, have a critical role in the development of epilepsy. In this study, we investigated the role of innate inflammation in an in vitro model of post-traumatic epileptogenesis. We combined organotypic hippocampal slice cultures, representing an in vitro model of post-traumatic epilepsy, with multi-electrode array recordings to directly monitor the development of epileptiform activity and to examine the concomitant changes in cytokine release, cell death, and glial cell activation. We report that synchronized ictal- and interictal-like activities spontaneously evolve in this culture. Dynamic changes in the release of the pro-inflammatory cytokines IL-1β, TNFα, and IL-6 were observed throughout the culture period (3 to 21 days in vitro) with persistent activation of microglia and astrocytes. We found that neutralizing TNFα with a polyclonal antibody significantly reduced ictal discharges, and this effect lasted for 1 week after antibody washout. Neither phenytoin nor an anti-IL-6 polyclonal antibody was efficacious in inhibiting the development of epileptiform activity. Our data show a sustained effect of the anti-TNFα antibody on the ictal progression in organotypic hippocampal slice cultures supporting the critical role of inflammatory mediators in epilepsy and establishing a proof-of-principle evidence for the utility of this preparation to test the therapeutic effects of anti-inflammatory treatments.
Collapse
Affiliation(s)
- Seon-Ah Chong
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium.
| | - Silvia Balosso
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | | | - Gregory Szczesny
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Etienne Hanon
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Kasper Claes
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Xavier Van Damme
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Bénédicte Danis
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Jonathan Van Eyll
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Christian Wolff
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | - Rafal M Kaminski
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | | |
Collapse
|
27
|
Chong SA, Balosso S, Vandenplas C, Szczesny G, Hanon E, Claes K, Van Damme X, Danis B, Van Eyll J, Wolff C, Vezzani A, Kaminski RM, Niespodziany I. Intrinsic Inflammation Is a Potential Anti-Epileptogenic Target in the Organotypic Hippocampal Slice Model. Neurotherapeutics 2018; 15:470-488. [PMID: 29464573 DOI: 10.1007/s13311-018-0607-6/figures/7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2023] Open
Abstract
Understanding the mechanisms of epileptogenesis is essential to develop novel drugs that could prevent or modify the disease. Neuroinflammation has been proposed as a promising target for therapeutic interventions to inhibit the epileptogenic process that evolves from traumatic brain injury. However, it remains unclear whether cytokine-related pathways, particularly TNFα signaling, have a critical role in the development of epilepsy. In this study, we investigated the role of innate inflammation in an in vitro model of post-traumatic epileptogenesis. We combined organotypic hippocampal slice cultures, representing an in vitro model of post-traumatic epilepsy, with multi-electrode array recordings to directly monitor the development of epileptiform activity and to examine the concomitant changes in cytokine release, cell death, and glial cell activation. We report that synchronized ictal- and interictal-like activities spontaneously evolve in this culture. Dynamic changes in the release of the pro-inflammatory cytokines IL-1β, TNFα, and IL-6 were observed throughout the culture period (3 to 21 days in vitro) with persistent activation of microglia and astrocytes. We found that neutralizing TNFα with a polyclonal antibody significantly reduced ictal discharges, and this effect lasted for 1 week after antibody washout. Neither phenytoin nor an anti-IL-6 polyclonal antibody was efficacious in inhibiting the development of epileptiform activity. Our data show a sustained effect of the anti-TNFα antibody on the ictal progression in organotypic hippocampal slice cultures supporting the critical role of inflammatory mediators in epilepsy and establishing a proof-of-principle evidence for the utility of this preparation to test the therapeutic effects of anti-inflammatory treatments.
Collapse
Affiliation(s)
- Seon-Ah Chong
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium.
| | - Silvia Balosso
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | | | - Gregory Szczesny
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Etienne Hanon
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Kasper Claes
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Xavier Van Damme
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Bénédicte Danis
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Jonathan Van Eyll
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Christian Wolff
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, 20156, Italy
| | - Rafal M Kaminski
- UCB Biopharma SPRL, Chemin du Foriest, B-1420, Braine l'Alleud, Belgium
| | | |
Collapse
|
28
|
Thompson R, Sakiyama-Elbert S. Using biomaterials to promote pro-regenerative glial phenotypes after nervous system injuries. ACTA ACUST UNITED AC 2018; 13:024104. [PMID: 29186011 DOI: 10.1088/1748-605x/aa9e23] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Trauma to either the central or peripheral nervous system (PNS) often leads to significant loss of function and disability in patients. This high rate of long-term disability is due to the overall limited regenerative potential of nervous tissue, even though the PNS has more regenerative potential than the central nervous system (CNS). The supporting glial cells in the periphery, Schwann cells, are part of the reason for the improved recovery observed in the PNS. In the CNS, the glial populations, astrocytes and oligodendrocytes (OLs), do not have as much potential to promote regeneration and are at times inhibitory to neuronal growth. In particular, the inhibitory roles astrocytes play following trauma has led to a historical focus on neurons and OLs instead of astrocytes. Recently, this focus has shifted as new, regenerative astrocyte phenotypes have been described. From these observations, glial cells clearly play critical roles in native recovery pathways in both the CNS and PNS. This makes the ability to manipulate both transplanted and native glial cell phenotypes a potentially successful strategy to improve nerve injury outcomes. This review focuses on factors that cause glial cells to adopt repair phenotypes and biomaterials that manipulate and/or harness these glial phenotypes.
Collapse
Affiliation(s)
- Russell Thompson
- Department of Biomedical Engineering, University of Texas at Austin 107 W Dean Keeton, Austin, TX 78712, United States of America. Department of Biomedical Engineering, Washington University in St. Louis, 1 Brooking Drive, St. Louis, MO 63130, United States of America
| | | |
Collapse
|
29
|
Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-Supported Cell Transplantation Treatments for Spinal Cord Injury: Challenges and Perspectives. Front Cell Neurosci 2018; 11:430. [PMID: 29375316 PMCID: PMC5768640 DOI: 10.3389/fncel.2017.00430] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
30
|
Katiyar KS, Winter CC, Gordián-Vélez WJ, O'Donnell JC, Song YJ, Hernandez NS, Struzyna LA, Cullen DK. Three-dimensional Tissue Engineered Aligned Astrocyte Networks to Recapitulate Developmental Mechanisms and Facilitate Nervous System Regeneration. J Vis Exp 2018. [PMID: 29364269 DOI: 10.3791/55848] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Neurotrauma and neurodegenerative disease often result in lasting neurological deficits due to the limited capacity of the central nervous system (CNS) to replace lost neurons and regenerate axonal pathways. However, during nervous system development, neuronal migration and axonal extension often occur along pathways formed by other cells, referred to as "living scaffolds". Seeking to emulate these mechanisms and to design a strategy that circumvents the inhibitory environment of the CNS, this manuscript presents a protocol to fabricate tissue engineered astrocyte-based "living scaffolds". To create these constructs, we employed a novel biomaterial encasement scheme to induce astrocytes to self-assemble into dense three-dimensional bundles of bipolar longitudinally-aligned somata and processes. First, hollow hydrogel micro-columns were assembled, and the inner lumen was coated with collagen extracellular-matrix. Dissociated cerebral cortical astrocytes were then delivered into the lumen of the cylindrical micro-column and, at a critical inner diameter of <350 µm, spontaneously self-aligned and contracted to produce long fiber-like cables consisting of dense bundles of astrocyte processes and collagen fibrils measuring <150 µm in diameter yet extending several cm in length. These engineered living scaffolds exhibited >97% cell viability and were virtually exclusively comprised of astrocytes expressing a combination of the intermediate filament proteins glial-fibrillary acidic protein (GFAP), vimentin, and nestin. These aligned astrocyte networks were found to provide a permissive substrate for neuronal attachment and aligned neurite extension. Moreover, these constructs maintain integrity and alignment when extracted from the hydrogel encasement, making them suitable for CNS implantation. These preformed constructs structurally emulate key cytoarchitectural elements of naturally occurring glial-based "living scaffolds" in vivo. As such, these engineered living scaffolds may serve as test-beds to study neurodevelopmental mechanisms in vitro or facilitate neuroregeneration by directing neuronal migration and/or axonal pathfinding following CNS degeneration in vivo.
Collapse
Affiliation(s)
- Kritika S Katiyar
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Michael J. Crescenz Veterans Affairs Medical Center; School of Biomedical Engineering, Drexel University
| | - Carla C Winter
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Michael J. Crescenz Veterans Affairs Medical Center; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania
| | - Wisberty J Gordián-Vélez
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Michael J. Crescenz Veterans Affairs Medical Center; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania
| | - John C O'Donnell
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Michael J. Crescenz Veterans Affairs Medical Center
| | - Yeri J Song
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania
| | - Nicole S Hernandez
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania
| | - Laura A Struzyna
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Michael J. Crescenz Veterans Affairs Medical Center; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania
| | - D Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania; Center for Neurotrauma, Neurodegeneration & Restoration, Michael J. Crescenz Veterans Affairs Medical Center; Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania;
| |
Collapse
|
31
|
Guarino V, Benfenati V, Cruz-Maya I, Saracino E, Zamboni R, Ambrosio L. Instructive proteins for tissue regeneration. FUNCTIONAL 3D TISSUE ENGINEERING SCAFFOLDS 2018:23-49. [DOI: 10.1016/b978-0-08-100979-6.00002-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
32
|
Urinary Tissue Engineering: Challenges and Opportunities. Sex Med Rev 2017; 6:35-44. [PMID: 29066225 DOI: 10.1016/j.sxmr.2017.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 01/14/2023]
Abstract
INTRODUCTION In this review, we discuss major advancements and common challenges in constructing and regenerating a neo-urinary conduit (NUC). First, we focus on the need for regenerating the urothelium, the hallmark the urine barrier, unique to urinary tissues. Second, we focus on clinically feasible scaffolds based on decellularized matrices and molded collagen that are currently of great research interest. AIM To discuss the major advancements in constructing a tissue-engineered NUC (TE-NUC) and the challenges involved in their successful clinical translation. METHODS A comprehensive search of peer-reviewed literature from PubMed and Google Scholar on subjects related to urothelium regeneration, decellularized tissue matrices, and collagen scaffolds was conducted. MAIN OUTCOME MEASURE We evaluated the main biological and mechanical functions of urinary tissues, the need for TE implants to create a urinary diversion, the reasons for their failures in clinical settings, and the applications of decellularized tissue matrices and collagen-based molded scaffolds in their regeneration. RESULTS It is necessary to create a urine barrier that prevents urine leakage into the stroma that can cause failure of the graft. Despite the regeneration potential of the urothelium, the limited supply of healthy urothelial cells in patients with bladder cancer remains a major challenge. In this context, alternative strategies, such as transdifferentiation of cells into urothelium or engineered scaffolds based on decellularized tissues and molded collagen with robust urine barrier properties, are active areas of research. CONCLUSION There is an immediate need for developing a functional TE-NUC that can improve the quality of life of patients with bladder cancer. It is possible to achieve a TE-NUC by bioengineering an implant that has appropriate biological and mechanical properties to store and transport urine. We anticipate that future advancements in urothelium regeneration and material design will lead us closer to successful neo-urinary tissue constructs. Singh A, Bivalacqua TJ, Sopko N. Urinary Tissue Engineering: Challenges and Opportunities. Sex Med Rev 2018;6:35-44.
Collapse
|
33
|
Abstract
The surface concentration gradient of two extracellular matrix (ECM) macromolecules was developed to study the migratory and morphological responses of astrocytes to molecular cues typically found in the central nervous system injury environment. The gradient, prepared using microcontact printing, was composed of randomly positioned micrometer-sized dots of aggrecan (AGG) printed on a substrate uniformly coated with laminin (LN). AGG dots were printed in an increasing number along the 1000 μm long and 50 μm wide gradient area which had on each end either a full surface coverage of AGG or LN. Each dot gradient was surrounded by a 100 μm-wide uniform field of AGG printed over laminin. Seeded astrocytes were found to predominantly attach to LN regions on the gradient. Cellular extensions of these cells were longer than the similar processes for cells seeded on uniform substrates of AGG or LN serving as controls. Astrocyte extensions were the largest and spanned a distance of 150 μm when the cells were attached to the mixed AGG+LN patches on the gradient. As evidenced by their increased area and perimeter, the cells extended processes in a stellate fashion upon initial attachment and maintained extensions when seeded in AGG+LN regions but not on uniform laminin controls. The cells migrated short distances, ∼20-35 μm, over 24 h and in doing so preferentially shifted from AGG areas to higher LN surface coverage regions. The results indicated that presenting mixed ECM cues caused astrocytes to sample larger areas of the substrate and made the cells to preferentially relocate to a more permissive ECM region.
Collapse
|
34
|
Antman-Passig M, Levy S, Gartenberg C, Schori H, Shefi O. Mechanically Oriented 3D Collagen Hydrogel for Directing Neurite Growth. Tissue Eng Part A 2017; 23:403-414. [PMID: 28437179 DOI: 10.1089/ten.tea.2016.0185] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent studies in the field of neuro-tissue engineering have demonstrated the promising effects of aligned contact guidance cue to scaffolds of enhancement and direction of neuronal growth. In vivo, neurons grow and develop neurites in a complex three-dimensional (3D) extracellular matrix (ECM) surrounding. Studies have utilized hydrogel scaffolds derived from ECM molecules to better simulate natural growth. While many efforts have been made to control neuronal growth on 2D surfaces, the development of 3D scaffolds with an elaborate oriented topography to direct neuronal growth still remains a challenge. In this study, we designed a method for growing neurons in an aligned and oriented 3D collagen hydrogel. We aligned collagen fibers by inducing controlled uniaxial strain on gels. To examine the collagen hydrogel as a suitable scaffold for neuronal growth, we evaluated the physical properties of the hydrogel and measured collagen fiber properties. By combining the neuronal culture in 3D collagen hydrogels with strain-induced alignment, we were able to direct neuronal growth in the direction of the aligned collagen matrix. Quantitative evaluation of neurite extension and directionality within aligned gels was performed. The analysis showed neurite growth aligned with collagen matrix orientation, while maintaining the advantageous 3D growth.
Collapse
Affiliation(s)
- Merav Antman-Passig
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Bar Ilan University , Ramat Gan, Israel
| | - Shahar Levy
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Bar Ilan University , Ramat Gan, Israel
| | - Chaim Gartenberg
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Bar Ilan University , Ramat Gan, Israel
| | - Hadas Schori
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Bar Ilan University , Ramat Gan, Israel
| | - Orit Shefi
- Faculty of Engineering, Bar Ilan Institute of Nanotechnologies and Advanced Materials, Bar Ilan University , Ramat Gan, Israel
| |
Collapse
|
35
|
Eberwein P, Reinhard T. [New biomaterials and alternative stem cell sources for the reconstruction of the limbal stem cell niche]. Ophthalmologe 2017; 114:318-326. [PMID: 28378048 DOI: 10.1007/s00347-017-0463-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Reconstruction of the limbal stem cell niche in patients with limbal stem cell insufficiency remains one of the most challenging tasks in the treatment of ocular surface diseases. Ex vivo expansion of limbal stem cells still has potential for optimization despite positive reports in centers worldwide. New biomaterials as well as alternative cell sources for the reconstruction of the limbal stem cell niche have been published in recent years. The aim of this review is to provide insight into new biomaterials and cell sources which may find their way into clinical routine in the upcoming decades.
Collapse
Affiliation(s)
- P Eberwein
- Klinik für Augenheilkunde, Uniklinikum Freiburg, Killianstr. 5, 79106, Freiburg, Deutschland.
| | - T Reinhard
- Klinik für Augenheilkunde, Uniklinikum Freiburg, Killianstr. 5, 79106, Freiburg, Deutschland
| |
Collapse
|
36
|
O'Rourke C, Lee-Reeves C, Drake RA, Cameron GW, Loughlin AJ, Phillips JB. Adapting tissue-engineered in vitro CNS models for high-throughput study of neurodegeneration. J Tissue Eng 2017; 8:2041731417697920. [PMID: 28507726 PMCID: PMC5415290 DOI: 10.1177/2041731417697920] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/14/2017] [Indexed: 11/21/2022] Open
Abstract
Neurodegenerative conditions remain difficult to treat, with the continuing failure to see therapeutic research successfully advance to clinical trials. One of the obstacles that must be overcome is to develop enhanced models of disease. Tissue engineering techniques enable us to create organised artificial central nervous system tissue that has the potential to improve the drug development process. This study presents a replicable model of neurodegenerative pathology through the use of engineered neural tissue co-cultures that can incorporate cells from various sources and allow degeneration and protection of neurons to be observed easily and measured, following exposure to neurotoxic compounds – okadaic acid and 1-methyl-4-phenylpyridinium. Furthermore, the technology has been miniaturised through development of a mould with 6 mm length that recreates the advantageous features of engineered neural tissue co-cultures at a scale suitable for commercial research and development. Integration of human-derived induced pluripotent stem cells aids more accurate modelling of human diseases, creating new possibilities for engineered neural tissue co-cultures and their use in drug screening.
Collapse
Affiliation(s)
- Caitriona O'Rourke
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK.,Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Charlotte Lee-Reeves
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | | | | | - A Jane Loughlin
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - James B Phillips
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| |
Collapse
|
37
|
Watson PMD, Kavanagh E, Allenby G, Vassey M. Bioengineered 3D Glial Cell Culture Systems and Applications for Neurodegeneration and Neuroinflammation. SLAS DISCOVERY 2017; 22:583-601. [PMID: 28346104 DOI: 10.1177/2472555217691450] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neurodegeneration and neuroinflammation are key features in a range of chronic central nervous system (CNS) diseases such as Alzheimer's and Parkinson's disease, as well as acute conditions like stroke and traumatic brain injury, for which there remains significant unmet clinical need. It is now well recognized that current cell culture methodologies are limited in their ability to recapitulate the cellular environment that is present in vivo, and there is a growing body of evidence to show that three-dimensional (3D) culture systems represent a more physiologically accurate model than traditional two-dimensional (2D) cultures. Given the complexity of the environment from which cells originate, and their various cell-cell and cell-matrix interactions, it is important to develop models that can be controlled and reproducible for drug discovery. 3D cell models have now been developed for almost all CNS cell types, including neurons, astrocytes, microglia, and oligodendrocyte cells. This review will highlight a number of current and emerging techniques for the culture of astrocytes and microglia, glial cell types with a critical role in neurodegenerative and neuroinflammatory conditions. We describe recent advances in glial cell culture using electrospun polymers and hydrogel macromolecules, and highlight how these novel culture environments influence astrocyte and microglial phenotypes in vitro, as compared to traditional 2D systems. These models will be explored to illuminate current trends in the techniques used to create 3D environments for application in research and drug discovery focused on astrocytes and microglial cells.
Collapse
|
38
|
Anisotropically organized three-dimensional culture platform for reconstruction of a hippocampal neural network. Nat Commun 2017; 8:14346. [PMID: 28146148 PMCID: PMC5296669 DOI: 10.1038/ncomms14346] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 12/19/2016] [Indexed: 01/06/2023] Open
Abstract
In native tissues, cellular and acellular components are anisotropically organized and often aligned in specific directions, providing structural and mechanical properties for actuating biological functions. Thus, engineering alignment not only allows for emulation of native tissue structures but might also enable implementation of specific functionalities. However, achieving desired alignment is challenging, especially in three-dimensional constructs. By exploiting the elastomeric property of polydimethylsiloxane and fibrillogenesis kinetics of collagen, here we introduce a simple yet effective method to assemble and align fibrous structures in a multi-modular three-dimensional conglomerate. Applying this method, we have reconstructed the CA3–CA1 hippocampal neural circuit three-dimensionally in a monolithic gel, in which CA3 neurons extend parallel axons to and synapse with CA1 neurons. Furthermore, we show that alignment of the fibrous scaffold facilitates the establishment of functional connectivity. This method can be applied for reconstructing other neural circuits or tissue units where anisotropic organization in a multi-modular structure is desired. Alignment or anisotropic organisation within and between cells enables biological function but is challenging to engineer. Here, the authors align collagen fibres in a pre-strained polydimethylsiloxane mould to generate a 3D scaffold that guides hippocampal neuron axon growth to form CA3–CA1 neural circuits.
Collapse
|
39
|
Sanen K, Martens W, Georgiou M, Ameloot M, Lambrichts I, Phillips J. Engineered neural tissue with Schwann cell differentiated human dental pulp stem cells: potential for peripheral nerve repair? J Tissue Eng Regen Med 2017; 11:3362-3372. [PMID: 28052540 DOI: 10.1002/term.2249] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 06/14/2016] [Accepted: 07/03/2016] [Indexed: 12/18/2022]
Abstract
Despite the spontaneous regenerative capacity of the peripheral nervous system, large gap peripheral nerve injuries (PNIs) require bridging strategies. The limitations and suboptimal results obtained with autografts or hollow nerve conduits in the clinic urge the need for alternative treatments. Recently, we have described promising neuroregenerative capacities of Schwann cells derived from differentiated human dental pulp stem cells (d-hDPSCs) in vitro. Here, we extended the in vitro assays to show the pro-angiogenic effects of d-hDPSCs, such as enhanced endothelial cell proliferation, migration and differentiation. In addition, for the first time we evaluated the performance of d-hDPSCs in an in vivo rat model of PNI. Eight weeks after transplantation of NeuraWrap™ conduits filled with engineered neural tissue (EngNT) containing aligned d-hDPSCs in 15-mm rat sciatic nerve defects, immunohistochemistry and ultrastructural analysis revealed ingrowing neurites, myelinated nerve fibres and blood vessels along the construct. Although further research is required to optimize the delivery of this EngNT, our findings suggest that d-hDPSCs are able to exert a positive effect in the regeneration of nerve tissue in vivo. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kathleen Sanen
- Biophysics Group, Biomedical Research Institute, Hasselt University, Agoralaan Building C, 3590, Diepenbeek, Belgium
| | - Wendy Martens
- Morphology Group, Biomedical Research Institute, Hasselt University, Agoralaan Building C, 3590, Diepenbeek, Belgium
| | - Melanie Georgiou
- Advanced Centre for Biochemical Engineering, University College London, Bernard Katz Building, Gordon Street, London, WC1H 0AH, UK
| | - Marcel Ameloot
- Biophysics Group, Biomedical Research Institute, Hasselt University, Agoralaan Building C, 3590, Diepenbeek, Belgium
| | - Ivo Lambrichts
- Morphology Group, Biomedical Research Institute, Hasselt University, Agoralaan Building C, 3590, Diepenbeek, Belgium
| | - James Phillips
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, 256 Gray's Inn Road, London, WC1X 8LD, UK
| |
Collapse
|
40
|
Yang W, Li L, Su G, Zhang Z, Cao Y, Li X, Shi Y, Zhang Q. A collagen telopeptide binding peptide shows potential in aiding collagen bundle formation and fibril orientation. Biomater Sci 2017. [DOI: 10.1039/c6bm00574h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A double-armed CTBP-PEG-CTBP derivative of a collagen telopeptide binding peptide (CTBP), shows potential in aiding collagen bundle formation and fibril orientation by interacting with fibrils.
Collapse
Affiliation(s)
- Wenyu Yang
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| | - Lin Li
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| | - Guanghao Su
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| | - Zhe Zhang
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| | - Yiting Cao
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| | - Xuemin Li
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| | - Yanping Shi
- School of Chemistry and Chemical Engineering
- Tianjin University of Technology
- Tianjin
- PR China
| | - Qiqing Zhang
- The Key Laboratory of Biomedical Material of Tianjin
- Institute of Biomedical Engineering
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Tianjin
| |
Collapse
|
41
|
Martínez GF, Bianchimano P, Brauer MM. Estrogen-induced collagen reorientation correlates with sympathetic denervation of the rat myometrium. Auton Neurosci 2016; 201:32-39. [PMID: 27639314 DOI: 10.1016/j.autneu.2016.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/08/2016] [Accepted: 09/09/2016] [Indexed: 12/16/2022]
Abstract
Estrogen inhibits the growth and causes the degeneration (pruning) of sympathetic nerves supplying the rat myometrium. Previous cryoculture studies evidenced that substrate-bound signals contribute to diminish the ability of the estrogenized myometrium to support sympathetic nerve growth. Using electron microscopy, here we examined neurite-substrate interactions in myometrial cryocultures, observing that neurites grew associated to collagen fibrils present in the surface of the underlying cryosection. In addition, we assessed quantitatively the effects of estrogen on myometrial collagen organization in situ, using ovariectomized rats treated with estrogen and immature females undergoing puberty. Under low estrogen levels, most collagen fibrils were oriented in parallel to the muscle long axis (83% and 85%, respectively). Following estrogen treatment, 89% of fibrils was oriented perpendicularly to the muscle main axis; while after puberty, 57% of fibrils acquired this orientation. Immunohistochemistry combined with histology revealed that the vast majority of fine sympathetic nerve fibers supplying the myometrium courses within the areas where collagen realignment was observed. Finally, to assess whether depending on their orientation collagen fibrils can promote or inhibit neurite outgrowth, we employed cryocultures, now using as substrate tissue sections of rat-tail tendon. We observed that neurites grew extensively in the direction of the parallel-aligned collagen fibrils in the tendon main axis but were inhibited to grow perpendicularly to this axis. Collectively, these findings support the hypothesis that collagen reorientation may be one of the factors contributing to diminish the neuritogenic capacity of the estrogen-primed myometrial substrate.
Collapse
Affiliation(s)
- G F Martínez
- Laboratory of Cell Biology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - P Bianchimano
- Laboratory of Cell Biology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - M M Brauer
- Laboratory of Cell Biology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
| |
Collapse
|
42
|
Takahashi H, Itoga K, Shimizu T, Yamato M, Okano T. Human Neural Tissue Construct Fabrication Based on Scaffold-Free Tissue Engineering. Adv Healthc Mater 2016; 5:1931-8. [PMID: 27331769 DOI: 10.1002/adhm.201600197] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/21/2016] [Indexed: 11/06/2022]
Abstract
Current neural tissue engineering strategies involve the development and application of neural tissue constructs produced by using an anisotropic polymeric scaffold. This study reports a scaffold-free method of tissue engineering to create a tubular neural tissue construct containing unidirectional neuron bundles. The surface patterning of a thermoresponsive culture substrate and a coculture system of neurons with patterned astrocytes can provide an anisotropic structure and easy handling of the neural tissue construct without the use of a scaffold. Furthermore, using a gelatin gel-coated plunger, the neuron bundles can be laid out in the same direction at regulated intervals within multilayered astrocyte sheets. Since the 3D tissue construct is composed only by neurons and astrocytes, they can communicate physiologically without obstruction of a scaffold. The medical benefits of scaffold-free tissue generation provide new opportunities for the development of human cell-based tissue models required to better understand the mechanisms of neurodegenerative diseases. Therefore, this new tissue engineering approach may be useful to establish a technology for regenerative medicine and drug discovery using the patient's own neurons.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; 8-1 Kawada-cho Shinjuku-ku Tokyo 162-8666 Japan
| | - Kazuyoshi Itoga
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; 8-1 Kawada-cho Shinjuku-ku Tokyo 162-8666 Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; 8-1 Kawada-cho Shinjuku-ku Tokyo 162-8666 Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; 8-1 Kawada-cho Shinjuku-ku Tokyo 162-8666 Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science; Tokyo Women's Medical University; 8-1 Kawada-cho Shinjuku-ku Tokyo 162-8666 Japan
| |
Collapse
|
43
|
Winter CC, Katiyar KS, Hernandez NS, Song YJ, Struzyna LA, Harris JP, Cullen DK. Transplantable living scaffolds comprised of micro-tissue engineered aligned astrocyte networks to facilitate central nervous system regeneration. Acta Biomater 2016; 38:44-58. [PMID: 27090594 DOI: 10.1016/j.actbio.2016.04.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/24/2016] [Accepted: 04/13/2016] [Indexed: 12/16/2022]
Abstract
UNLABELLED Neurotrauma, stroke, and neurodegenerative disease may result in widespread loss of neural cells as well as the complex interconnectivity necessary for proper central nervous system function, generally resulting in permanent functional deficits. Potential regenerative strategies involve the recruitment of endogenous neural stem cells and/or directed axonal regeneration through the use of tissue engineered "living scaffolds" built to mimic features of three-dimensional (3-D) in vivo migratory or guidance pathways. Accordingly, we devised a novel biomaterial encasement scheme using tubular hydrogel-collagen micro-columns that facilitated the self-assembly of seeded astrocytes into 3-D living scaffolds consisting of long, cable-like aligned astrocytic networks. Here, robust astrocyte alignment was achieved within a micro-column inner diameter (ID) of 180μm or 300-350μm but not 1.0mm, suggesting that radius of curvature dictated the extent of alignment. Moreover, within small ID micro-columns, >70% of the astrocytes assumed a bi-polar morphology, versus ∼10% in larger micro-columns or planar surfaces. Cell-cell interactions also influenced the aligned architecture, as extensive astrocyte-collagen contraction was achieved at high (9-12×10(5)cells/mL) but not lower (2-6×10(5)cells/mL) seeding densities. This high density micro-column seeding led to the formation of ultra-dense 3-D "bundles" of aligned bi-polar astrocytes within collagen measuring up to 150μm in diameter yet extending to a remarkable length of over 2.5cm. Importantly, co-seeded neurons extended neurites directly along the aligned astrocytic bundles, demonstrating permissive cues for neurite extension. These transplantable cable-like astrocytic networks structurally mimic the glial tube that guides neuronal progenitor migration in vivo along the rostral migratory stream, and therefore may be useful to guide progenitor cells to repopulate sites of widespread neurodegeneration. STATEMENT OF SIGNIFICANCE This manuscript details our development of novel micro-tissue engineering techniques to generate robust networks of longitudinally aligned astrocytes within transplantable micro-column hydrogels. We report a novel biomaterial encasement scheme that facilitated the self-assembly of seeded astrocytes into long, aligned regenerative pathways. These miniature "living scaffold" constructs physically emulate the glial tube - a pathway in the brain consisting of aligned astrocytes that guide the migration of neuronal progenitor cells - and therefore may facilitate directed neuronal migration for central nervous system repair. The small size and self-contained design of these aligned astrocyte constructs will permit minimally invasive transplantation in models of central nervous system injury in future studies.
Collapse
|
44
|
Silver J. The glial scar is more than just astrocytes. Exp Neurol 2016; 286:147-149. [PMID: 27328838 DOI: 10.1016/j.expneurol.2016.06.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Jerry Silver
- Case Western Reserve University, School of Medicine, Department of Neurosciences, Cleveland, OH 44106, USA.
| |
Collapse
|
45
|
Katiyar KS, Winter CC, Struzyna LA, Harris JP, Cullen DK. Mechanical elongation of astrocyte processes to create living scaffolds for nervous system regeneration. J Tissue Eng Regen Med 2016; 11:2737-2751. [PMID: 27273796 DOI: 10.1002/term.2168] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 12/31/2015] [Accepted: 02/03/2016] [Indexed: 12/21/2022]
Abstract
Following brain injury or neurodegenerative disease, successful regeneration requires orchestrated migration of neurons and reformation of long-distance communication fibres, or axons. Such extensive regeneration does not occur in the mature brain; however, during embryonic development, pathways formed by glial cells extend several millimeters (mm) to create 'living scaffolds' for targeted neural cell migration and axonal pathfinding. Techniques to recapitulate long process outgrowth in glial cells have proven elusive, preventing the exploitation of this developmental mechanism for regeneration. In the current study, astrocytes were induced to form a network of interconnected processes that were subjected to controlled mechanical tension in vitro using custom-built mechanobioreactors. We discovered a specific micron (μm)-scale mechanical growth regime that induced elongation of the astrocytic processes to a remarkable length of 2.5 mm at an optimal rate of 12.5 μm/h. More rapid mechanical regimes (> 20 μm/h) caused greater incidence of process degeneration or outright breakage, whereas slow regimes (< 4 μm/h) led to adaptive motility, thus failing to achieve process elongation. Cellular phenotype for this astrocytic 'stretch-growth' was confirmed based on presentation of the intermediate filament glial fibrillary acidic protein (GFAP). Mechanical elongation resulted in the formation of dense bundles of aligned astrocytic processes. Importantly, seeded neurons readily adhered to, and extended neurites directly along, the elongated astrocytic processes, demonstrating permissiveness to support neuronal growth. This is the first demonstration of the controlled application of mechanical forces to create long astrocytic processes, which may form the backbone of tissue-engineered 'living scaffolds' that structurally emulate radial glia to facilitate neuroregeneration. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kritika S Katiyar
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,School of Biomedical Engineering, Drexel University, Philadelphia, PA, USA
| | - Carla C Winter
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura A Struzyna
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - James P Harris
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - D Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| |
Collapse
|
46
|
Al-Ali H, Beckerman SR, Bixby JL, Lemmon VP. In vitro models of axon regeneration. Exp Neurol 2016; 287:423-434. [PMID: 26826447 DOI: 10.1016/j.expneurol.2016.01.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/20/2016] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
A variety of in vitro models have been developed to understand the mechanisms underlying the regenerative failure of central nervous system (CNS) axons, and to guide pre-clinical development of regeneration-promoting therapeutics. These range from single-cell based assays that typically focus on molecular mechanisms to organotypic assays that aim to recapitulate in vivo behavior. By utilizing a combination of models, researchers can balance the speed, convenience, and mechanistic resolution of simpler models with the biological relevance of more complex models. This review will discuss a number of models that have been used to build our understanding of the molecular mechanisms of CNS axon regeneration.
Collapse
Affiliation(s)
- Hassan Al-Ali
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Samuel R Beckerman
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Molecular & Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Center for Computational Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
47
|
Levis HJ, Daniels JT. Recreating the Human Limbal Epithelial Stem Cell Niche with Bioengineered Limbal Crypts. Curr Eye Res 2016; 41:1153-60. [PMID: 26727236 DOI: 10.3109/02713683.2015.1095932] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Creation of an in vitro model incorporating specific features that characterize a particular stem niche would allow researchers to study stem cell behavior in a more physiological environment. MATERIALS AND METHODS We have developed a tissue engineering process (RAFT) that rapidly and reliably creates bioengineered limbal crypts (BLCs) in the surface of collagen-based tissue equivalents (TEs). These BLCs mimic the three-dimensional topography of the limbal crypts (LCs), located in the limbal region of the human cornea, which are home to a population of limbal epithelial stem cells (LESCs). RESULTS Human limbal epithelial (hLE) cells occupying our BLCs expressed putative LESC markers such as ΔNp63α and Bmi1 and produced basement membrane proteins such as laminin β1 and laminin γ3; expression patterns are very similar to those seen in native LCs. Human limbal stromal cells elongate and align along the edge of native LCs and in our RAFT TEs, human limbal fibroblasts (hLFs) also appeared to exhibit this alignment and elongation behavior in response to the BLC topography. CONCLUSIONS We have demonstrated that we can maintain an immature population of hLE cells and aligned stromal cells in our BLCs to mimic some elements of the complexity of the human LESC niche.
Collapse
Affiliation(s)
- Hannah J Levis
- a Department of Ocular Biology and Therapeutics , UCL Institute of Ophthalmology , London , UK
| | - Julie T Daniels
- a Department of Ocular Biology and Therapeutics , UCL Institute of Ophthalmology , London , UK
| |
Collapse
|
48
|
Gonzalez-Andrades M, Cardona JDLC, Ionescu AM, Mosse CA, Brown RA. Photographic-Based Optical Evaluation of Tissues and Biomaterials Used for Corneal Surface Repair: A New Easy-Applied Method. PLoS One 2015; 10:e0142099. [PMID: 26566050 PMCID: PMC4643926 DOI: 10.1371/journal.pone.0142099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/16/2015] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Tissues and biomaterials used for corneal surface repair require fulfilling specific optical standards prior to implantation in the patient. However, there is not a feasible evaluation method to be applied in clinical or Good Manufacturing Practice settings. In this study, we describe and assess an innovative easy-applied photographic-based method (PBM) for measuring functional optical blurring and transparency in corneal surface grafts. METHODS Plastic compressed collagen scaffolds (PCCS) and multilayered amniotic membranes (AM) samples were optically and histologically evaluated. Transparency and image blurring measures were obtained by PBM, analyzing photographic images of a standardized band pattern taken through the samples. These measures were compared and correlated to those obtained applying the Inverse Adding-Doubling (IAD) technique, which is the gold standard method. RESULTS All the samples used for optical evaluation by PBM or IAD were histological suitable. PCCS samples presented transmittance values higher than 60%, values that increased with increasing wavelength as determined by IAD. The PBM indicated that PCCS had a transparency ratio (TR) value of 80.3 ± 2.8%, with a blurring index (BI) of 50.6 ± 4.2%. TR and BI obtained from the PBM showed a high correlation (ρ>|0.6|) with the diffuse transmittance and the diffuse reflectance, both determined using the IAD (p<0.005). The AM optical properties showed that there was a largely linear relationship between the blurring and the number of amnion layers, with more layers producing greater blurring. CONCLUSIONS This innovative proposed method represents an easy-applied technique for evaluating transparency and blurriness of tissues and biomaterials used for corneal surface repair.
Collapse
Affiliation(s)
- Miguel Gonzalez-Andrades
- Tissue Repair & Engineering Centre, University College of London, London, United Kingdom
- Tissue Engineering Group, Department of Histology, University of Granada, Granada, Spain
- Ophthalmology Department, San Cecilio University Hospital, Granada, Spain
- * E-mail:
| | - Juan de la Cruz Cardona
- Laboratory of Biomaterials and Optics—Optics Department, University of Granada, Granada, Spain
| | - Ana Maria Ionescu
- Laboratory of Biomaterials and Optics—Optics Department, University of Granada, Granada, Spain
| | - Charles A. Mosse
- Optics Department, University College of London, London, United Kingdom
| | - Robert A. Brown
- Tissue Repair & Engineering Centre, University College of London, London, United Kingdom
| |
Collapse
|
49
|
Struzyna LA, Wolf JA, Mietus CJ, Adewole DO, Chen HI, Smith DH, Cullen DK. Rebuilding Brain Circuitry with Living Micro-Tissue Engineered Neural Networks. Tissue Eng Part A 2015; 21:2744-56. [PMID: 26414439 DOI: 10.1089/ten.tea.2014.0557] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Prominent neuropathology following trauma, stroke, and various neurodegenerative diseases includes neuronal degeneration as well as loss of long-distance axonal connections. While cell replacement and axonal pathfinding strategies are often explored independently, there is no strategy capable of simultaneously replacing lost neurons and re-establishing long-distance axonal connections in the central nervous system. Accordingly, we have created micro-tissue engineered neural networks (micro-TENNs), which are preformed constructs consisting of long integrated axonal tracts spanning discrete neuronal populations. These living micro-TENNs reconstitute the architecture of long-distance axonal tracts, and thus may serve as an effective substrate for targeted neurosurgical reconstruction of damaged pathways in the brain. Cerebral cortical neurons or dorsal root ganglia neurons were precisely delivered into the tubular constructs, and properties of the hydrogel exterior and extracellular matrix internal column (180-500 μm diameter) were optimized for robust neuronal survival and to promote axonal extensions across the 2.0 cm tube length. The very small diameter permits minimally invasive delivery into the brain. In this study, preformed micro-TENNs were stereotaxically injected into naive rats to bridge deep thalamic structures with the cerebral cortex to assess construct survival and integration. We found that micro-TENN neurons survived at least 1 month and maintained their long axonal architecture along the cortical-thalamic axis. Notably, we also found neurite penetration from micro-TENN neurons into the host cortex, with evidence of synapse formation. These micro-TENNs represent a new strategy to facilitate nervous system repair by recapitulating features of neural pathways to restore or modulate damaged brain circuitry.
Collapse
Affiliation(s)
- Laura A Struzyna
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Philadelphia Veterans Affairs Medical Center , Philadelphia, Pennsylvania.,3 Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania , Philadelphia, Pennsylvania
| | - John A Wolf
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Philadelphia Veterans Affairs Medical Center , Philadelphia, Pennsylvania
| | - Constance J Mietus
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Dayo O Adewole
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Philadelphia Veterans Affairs Medical Center , Philadelphia, Pennsylvania.,3 Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania , Philadelphia, Pennsylvania
| | - H Isaac Chen
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Philadelphia Veterans Affairs Medical Center , Philadelphia, Pennsylvania
| | - Douglas H Smith
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - D Kacy Cullen
- 1 Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Philadelphia Veterans Affairs Medical Center , Philadelphia, Pennsylvania
| |
Collapse
|
50
|
Chwalek K, Sood D, Cantley WL, White JD, Tang-Schomer M, Kaplan DL. Engineered 3D Silk-collagen-based Model of Polarized Neural Tissue. J Vis Exp 2015:e52970. [PMID: 26555926 PMCID: PMC4692668 DOI: 10.3791/52970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Despite huge efforts to decipher the anatomy, composition and function of the brain, it remains the least understood organ of the human body. To gain a deeper comprehension of the neural system scientists aim to simplistically reconstruct the tissue by assembling it in vitro from basic building blocks using a tissue engineering approach. Our group developed a tissue-engineered silk and collagen-based 3D brain-like model resembling the white and gray matter of the cortex. The model consists of silk porous sponge, which is pre-seeded with rat brain-derived neurons, immersed in soft collagen matrix. Polarized neuronal outgrowth and network formation is observed with separate axonal and cell body localization. This compartmental architecture allows for the unique development of niches mimicking native neural tissue, thus enabling research on neuronal network assembly, axonal guidance, cell-cell and cell-matrix interactions and electrical functions.
Collapse
Affiliation(s)
| | - Disha Sood
- Department of Biomedical Engineering, Tufts University
| | | | - James D White
- Department of Biomedical Engineering, Tufts University
| | - Min Tang-Schomer
- Department of Pediatrics, University of Connecticut Health Center & Connecticut Children's Medical Center
| | | |
Collapse
|