1
|
Sarma S, Sudarshan TR, Nguyen V, Robang AS, Xiao X, Le JV, Helmicki ME, Paravastu AK, Hall CK. Design of parallel 𝛽-sheet nanofibrils using Monte Carlo search, coarse-grained simulations, and experimental testing. Protein Sci 2024; 33:e5102. [PMID: 39037281 PMCID: PMC11261811 DOI: 10.1002/pro.5102] [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: 11/10/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/23/2024]
Abstract
Peptide self-assembly into amyloid fibrils provides numerous applications in drug delivery and biomedical engineering applications. We augment our previously-established computational screening technique along with experimental biophysical characterization to discover 7-mer peptides that self-assemble into "parallel β-sheets", that is, β-sheets with N-terminus-to-C-terminus 𝛽-strand vectors oriented in parallel. To accomplish the desired β-strand organization, we applied the PepAD amino acid sequence design software to the Class-1 cross-β spine defined by Sawaya et al. This molecular configuration includes two layers of parallel β-sheets stacked such that N-terminus-to-C-terminus vectors are oriented antiparallel for molecules on adjacent β-sheets. The first cohort of PepAD identified peptides were examined for their fibrillation behavior in DMD/PRIME20 simulations, and the top performing sequence was selected as a prototype for a subsequent round of sequence refinement. The two rounds of design resulted in a library of eight 7-mer peptides. In DMD/PRIME20 simulations, five of these peptides spontaneously formed fibril-like structures with a predominantly parallel 𝛽-sheet arrangement, two formed fibril-like structure with <50% in parallel 𝛽-sheet arrangement and one remained a random coil. Among the eight candidate peptides produced by PepAD and DMD/PRIME20, five were synthesized and purified. All five assembled into amyloid fibrils composed of parallel β-sheets based on Fourier transform infrared spectroscopy, circular dichroism, electron microscopy, and thioflavin-T fluorescence spectroscopy measurements.
Collapse
Affiliation(s)
- Sudeep Sarma
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Tarunya Rao Sudarshan
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Van Nguyen
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Alicia S. Robang
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Xingqing Xiao
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
- Present address:
Department of Chemistry, School of Chemistry and Chemical EngineeringHainan UniversityHaikou CityHainan ProvincePeople's Republic of China
| | - Justin V. Le
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Michael E. Helmicki
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Anant K. Paravastu
- Department of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Carol K. Hall
- Department of Chemical and Biomolecular EngineeringNorth Carolina State UniversityRaleighNorth CarolinaUSA
| |
Collapse
|
2
|
Jafari A. Advancements in self-assembling peptides: Bridging gaps in 3D cell culture and electronic device fabrication. J Biomater Appl 2024; 38:1013-1035. [PMID: 38502905 PMCID: PMC11055414 DOI: 10.1177/08853282241240139] [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: 03/21/2024]
Abstract
Self-assembling peptides (SAPs) show promise in creating synthetic microenvironments that regulate cellular function and tissue repair. Also, the precise π-π interactions and hydrogen bonding within self-assembled peptide structures enable the creation of quantum confined structures, leading to reduced band gaps and the emergence of semiconductor properties within the superstructures. This review emphasizes the need for standardized 3D cell culture methods and electronic devices based on SAPs for monitoring cell communication and controlling cell surface morphology. Additionally, the gap in understanding the relationship between SAP peptide sequences and nanostructures is highlighted, underscoring the importance of optimizing peptide deposition parameters, which affect charge transport and bioactivity due to varying morphologies. The potential of peptide nanofibers as extracellular matrix mimics and the introduction of the zone casting method for improved film deposition are discussed within this review, aiming to bridge knowledge gaps and offer insights into fields like tissue engineering and materials science, with the potential for groundbreaking applications at the interface of biology and materials engineering.
Collapse
Affiliation(s)
- Azadeh Jafari
- Faculty of Applied Sciences, Simon Fraser University, Surrey, BC, Canada
| |
Collapse
|
3
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
5
|
Sharick JT, Atieh AJ, Gooch KJ, Leight JL. Click chemistry functionalization of self-assembling peptide hydrogels. J Biomed Mater Res A 2023; 111:389-403. [PMID: 36210776 PMCID: PMC10092743 DOI: 10.1002/jbm.a.37460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 01/12/2023]
Abstract
Self-assembling peptide (SAP) hydrogels provide a fibrous microenvironment to cells while also giving users control of biochemical and mechanical cues. Previously, biochemical cues were introduced by physically mixing them with SAPs prior to hydrogel assembly, or by incorporating them into the SAP sequence during peptide synthesis, which limited flexibility and increased costs. To circumvent these limitations, we developed "Click SAPs," a novel formulation that can be easily functionalized via click chemistry thiol-ene reaction. Due to its high cytocompatibility, the thiol-ene click reaction is currently used to crosslink and functionalize other types of polymeric hydrogels. In this study, we developed a click chemistry compatible SAP platform by addition of a modified lysine (lysine-alloc) to the SAP sequence, enabling effective coupling of thiol-containing molecules to the SAP hydrogel network. We demonstrate the flexibility of this approach by incorporating a fluorescent dye, a cellular adhesion peptide, and a matrix metalloproteinase-sensitive biosensor using the thiol-ene reaction in 3D Click SAPs. Using atomic force microscopy, we demonstrate that Click SAPs retain the ability to self-assemble into fibers, similar to previous systems. Additionally, a range of physiologically relevant stiffnesses can be achieved by adjusting SAP concentration. Encapsulated cells maintain high viability in Click SAPs and can interact with adhesion peptides and a matrix metalloproteinase biosensor, demonstrating that incorporated molecules retain their biological activity. The Click SAP platform supports easier functionalization with a wider array of bioactive molecules and enables new investigations with temporal and spatial control of the cellular microenvironment.
Collapse
Affiliation(s)
- Joe T Sharick
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.,The Center for Cancer Engineering, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Angelina J Atieh
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.,The Center for Cancer Engineering, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.,Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer L Leight
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.,The Center for Cancer Engineering, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
6
|
Sun W, Gregory DA, Zhao X. Designed peptide amphiphiles as scaffolds for tissue engineering. Adv Colloid Interface Sci 2023; 314:102866. [PMID: 36898186 DOI: 10.1016/j.cis.2023.102866] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023]
Abstract
Peptide amphiphiles (PAs) are peptide-based molecules that contain a peptide sequence as a head group covalently conjugated to a hydrophobic segment, such as lipid tails. They can self-assemble into well-ordered supramolecular nanostructures such as micelles, vesicles, twisted ribbons and nanofibers. In addition, the diversity of natural amino acids gives the possibility to produce PAs with different sequences. These properties along with their biocompatibility, biodegradability and a high resemblance to native extracellular matrix (ECM) have resulted in PAs being considered as ideal scaffold materials for tissue engineering (TE) applications. This review introduces the 20 natural canonical amino acids as building blocks followed by highlighting the three categories of PAs: amphiphilic peptides, lipidated peptide amphiphiles and supramolecular peptide amphiphile conjugates, as well as their design rules that dictate the peptide self-assembly process. Furthermore, 3D bio-fabrication strategies of PAs hydrogels are discussed and the recent advances of PA-based scaffolds in TE with the emphasis on bone, cartilage and neural tissue regeneration both in vitro and in vivo are considered. Finally, future prospects and challenges are discussed.
Collapse
Affiliation(s)
- Weizhen Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - David Alexander Gregory
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK; Department of Material Science and Engineering, University of Sheffield, Sheffield S3 7HQ, UK
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
7
|
Sung TC, Wang T, Liu Q, Ling QD, Subbiah SK, Renuka RR, Hsu ST, Umezawa A, Higuchi A. Cell-binding peptides on the material surface guide stem cell fate of adhesion, proliferation and differentiation. J Mater Chem B 2023; 11:1389-1415. [PMID: 36727243 DOI: 10.1039/d2tb02601e] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human cells, especially stem cells, need to communicate and interact with extracellular matrix (ECM) proteins, which not only serve as structural components but also guide and support cell fate and properties such as cell adhesion, proliferation, survival and differentiation. The binding of the cells with ECM proteins or ECM-derived peptides via cell adhesion receptors such as integrins activates several signaling pathways that determine the cell fate, morphological change, proliferation and differentiation. The development of synthetic ECM protein-derived peptides that mimic the biological and biochemical functions of natural ECM proteins will benefit academic and clinical application. Peptides derived from or inspired by specific ECM proteins can act as agonists of each ECM protein receptor. Given that most ECM proteins function in cell adhesion via integrin receptors, many peptides have been developed that bind to specific integrin receptors. In this review, we discuss the peptide sequence, immobilization design, reaction method, and functions of several ECM protein-derived peptides. Various peptide sequences derived from mainly ECM proteins, which are used for coating or grafting on dishes, scaffolds, hydrogels, implants or nanofibers, have been developed to improve the adhesion, proliferation or differentiation of stem cells and to culture differentiated cells. This review article will help to inform the optimal choice of ECM protein-derived peptides for the development of scaffolds, implants, hydrogels, nanofibers and 2D cell culture dishes to regulate the proliferation and direct the differentiation of stem cells into specific lineages.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qian Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Hsi-Chi City, Taipei 221, Taiwan
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Tambaram East, Chennai-73, 600078, India
| | - Remya Rajan Renuka
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Tambaram East, Chennai-73, 600078, India
| | - Shih-Tien Hsu
- Department of Internal Medicine, Taiwan Landseed Hospital, 77 Kuangtai Road, Pingjen City, Tao-Yuan County 32405, Taiwan
| | - Akihiro Umezawa
- Department of Reproduction, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China. .,Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan. .,R & D Center for Membrane Technology, Chung Yuan Christian University, 200 Chung-Bei Rd., Jhongli, Taoyuan 320, Taiwan
| |
Collapse
|
8
|
Martinez B, Peplow PV. Biomaterial and tissue-engineering strategies for the treatment of brain neurodegeneration. Neural Regen Res 2022; 17:2108-2116. [PMID: 35259816 PMCID: PMC9083174 DOI: 10.4103/1673-5374.336132] [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] [Indexed: 11/27/2022] Open
Abstract
The incidence of neurodegenerative diseases is increasing due to changing age demographics and the incidence of sports-related traumatic brain injury is tending to increase over time. Currently approved medicines for neurodegenerative diseases only temporarily reduce the symptoms but cannot cure or delay disease progression. Cell transplantation strategies offer an alternative approach to facilitating central nervous system repair, but efficacy is limited by low in vivo survival rates of cells that are injected in suspension. Transplanting cells that are attached to or encapsulated within a suitable biomaterial construct has the advantage of enhancing cell survival in vivo. A variety of biomaterials have been used to make constructs in different types that included nanoparticles, nanotubes, microspheres, microscale fibrous scaffolds, as well as scaffolds made of gels and in the form of micro-columns. Among these, Tween 80-methoxy poly(ethylene glycol)-poly(lactic-co-glycolic acid) nanoparticles loaded with rhynchophylline had higher transport across a blood-brain barrier model and decreased cell death in an in vitro model of Alzheimer’s disease than rhynchophylline or untreated nanoparticles with rhynchophylline. In an in vitro model of Parkinson’s disease, trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin had a similar protective ability as free non-Fe hemin. A positive effect on neuron survival in several in vivo models of Parkinson’s disease was associated with the use of biomaterial constructs such as trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin, carbon nanotubes with olfactory bulb stem cells, poly(lactic-co-glycolic acid) microspheres with attached DI-MIAMI cells, ventral midbrain neurons mixed with short fibers of poly-(L-lactic acid) scaffolds and reacted with xyloglucan with/without glial-derived neurotrophic factor, ventral midbrain neurons mixed with Fmoc-DIKVAV hydrogel with/without glial-derived neurotrophic factor. Further studies with in vivo models of Alzheimer’s disease and Parkinson’s disease are warranted especially using transplantation of cells in agarose micro-columns with an inner lumen filled with an appropriate extracellular matrix material.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
9
|
Bhat SM, Badiger VA, Vasishta S, Chakraborty J, Prasad S, Ghosh S, Joshi MB. 3D tumor angiogenesis models: recent advances and challenges. J Cancer Res Clin Oncol 2021; 147:3477-3494. [PMID: 34613483 PMCID: PMC8557138 DOI: 10.1007/s00432-021-03814-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/21/2021] [Indexed: 01/02/2023]
Abstract
The development of blood vessels, referred to as angiogenesis, is an intricate process regulated spatially and temporally through a delicate balance between the qualitative and quantitative expression of pro and anti-angiogenic molecules. As angiogenesis is a prerequisite for solid tumors to grow and metastasize, a variety of tumor angiogenesis models have been formulated to better understand the underlying mechanisms and associated clinical applications. Studies have demonstrated independent mechanisms inducing angiogenesis in tumors such as (a) HIF-1/VEGF mediated paracrine interactions between a cancer cell and endothelial cells, (b) recruitment of progenitor endothelial cells, and (c) vasculogenic mimicry. Moreover, single-cell sequencing technologies have indicated endothelial cell heterogeneity among organ systems including tumor tissues. However, existing angiogenesis models often rely upon normal endothelial cells which significantly differ from tumor endothelial cells exhibiting distinct (epi)genetic and metabolic signatures. Besides, the existence of intra-individual variations necessitates the development of improved tumor vascular model systems for personalized medicine. In the present review, we summarize recent advancements of 3D tumor vascular model systems which include (a) tissue engineering-based tumor models; (b) vascular organoid models, and (c) organ-on-chips and their importance in replicating the tumor angiogenesis along with the associated challenges to design improved models.
Collapse
Affiliation(s)
- Sharath M Bhat
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Vaishnavi A Badiger
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Juhi Chakraborty
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Seetharam Prasad
- Department of Surgery, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
10
|
Peressotti S, Koehl GE, Goding JA, Green RA. Self-Assembling Hydrogel Structures for Neural Tissue Repair. ACS Biomater Sci Eng 2021; 7:4136-4163. [PMID: 33780230 PMCID: PMC8441975 DOI: 10.1021/acsbiomaterials.1c00030] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
Hydrogel materials have been employed as biological scaffolds for tissue regeneration across a wide range of applications. Their versatility and biomimetic properties make them an optimal choice for treating the complex and delicate milieu of neural tissue damage. Aside from finely tailored hydrogel properties, which aim to mimic healthy physiological tissue, a minimally invasive delivery method is essential to prevent off-target and surgery-related complications. The specific class of injectable hydrogels termed self-assembling peptides (SAPs), provide an ideal combination of in situ polymerization combined with versatility for biofunctionlization, tunable physicochemical properties, and high cytocompatibility. This review identifies design criteria for neural scaffolds based upon key cellular interactions with the neural extracellular matrix (ECM), with emphasis on aspects that are reproducible in a biomaterial environment. Examples of the most recent SAPs and modification methods are presented, with a focus on biological, mechanical, and topographical cues. Furthermore, SAP electrical properties and methods to provide appropriate electrical and electrochemical cues are widely discussed, in light of the endogenous electrical activity of neural tissue as well as the clinical effectiveness of stimulation treatments. Recent applications of SAP materials in neural repair and electrical stimulation therapies are highlighted, identifying research gaps in the field of hydrogels for neural regeneration.
Collapse
Affiliation(s)
- Sofia Peressotti
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Gillian E. Koehl
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Josef A. Goding
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| | - Rylie A. Green
- Department
of Bioengineering and Centre for Neurotechnology, Imperial College London, London SW72AS, United Kingdom
| |
Collapse
|
11
|
Marchini A, Gelain F. Synthetic scaffolds for 3D cell cultures and organoids: applications in regenerative medicine. Crit Rev Biotechnol 2021; 42:468-486. [PMID: 34187261 DOI: 10.1080/07388551.2021.1932716] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Three-dimensional (3D) cell cultures offer an unparalleled platform to recreate spatial arrangements of cells in vitro. 3D cell culture systems have gained increasing interest due to their evident advantages in providing more physiologically relevant information and more predictive data compared to their two-dimensional (2D) counterpart. Design and well-established fabrication of organoids (a particular type of 3D cell culture system) are nowadays considered a pivotal achievement for their ability to replicate in vitro cytoarchitecture and the functionalities of an organ. In this condition, pluripotent stem cells self-organize into 3D structures mimicking the in vivo microenvironments, architectures and multi-lineage differentiation. Scaffolds are key supporting elements in these 3D constructs, and Matrigel is the most commonly used matrix despite its relevant translation limitations including animal-derived sources, non-defined composition, batch-to-batch variability and poorly tailorable properties. Alternatively, 3D synthetic scaffolds, including self-assembling peptides, show promising biocompatibility and biomimetic properties, and can be tailored on specific target tissue/cells. In this review, we discuss the recent advances on 3D cell culture systems and organoids, promising tools for tissue engineering and regenerative medicine applications. For this purpose, we will describe the current state-of-the-art on 3D cell culture systems and organoids based on currently available synthetic-based biomaterials (including tailored self-assembling peptides) either tested in in vivo animal models or developed in vitro with potential application in the field of tissue engineering, with the aim to inspire researchers to take on such promising platforms for clinical applications in the near future.
Collapse
Affiliation(s)
- Amanda Marchini
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Fabrizio Gelain
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
12
|
Sharma P, Pal VK, Roy S. An overview of latest advances in exploring bioactive peptide hydrogels for neural tissue engineering. Biomater Sci 2021; 9:3911-3938. [PMID: 33973582 DOI: 10.1039/d0bm02049d] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural tissue engineering holds great potential in addressing current challenges faced by medical therapies employed for the functional recovery of the brain. In this context, self-assembling peptides have gained considerable interest owing to their diverse physicochemical properties, which enable them to closely mimic the biophysical characteristics of the native ECM. Additionally, in contrast to synthetic polymers, which lack inherent biological signaling, peptide-based nanomaterials could be easily designed to present essential biological cues to the cells to promote cellular adhesion. Moreover, injectability of these biomaterials further widens their scope in biomedicine. In this context, hydrogels obtained from short bioactive peptide sequences are of particular interest owing to their facile synthesis and highly tunable properties. In spite of their well-known advantages, the exploration of short peptides for neural tissue engineering is still in its infancy and thus detailed discussion is required to evoke interest in this direction. This review provides a general overview of various bioactive hydrogels derived from short peptide sequences explored for neural tissue engineering. The review also discusses the current challenges in translating the benefits of these hydrogels to clinical practices and presents future perspectives regarding the utilization of these hydrogels for advanced biomedical applications.
Collapse
Affiliation(s)
- Pooja Sharma
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| | - Vijay Kumar Pal
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| | - Sangita Roy
- Institute of Nano Science and Technology, Sector 81, Knowledge city, Mohali, 140306, Punjab, India.
| |
Collapse
|
13
|
Distaffen HE, Jones CW, Abraham BL, Nilsson BL. Multivalent display of chemical signals on
self‐assembled
peptide scaffolds. Pept Sci (Hoboken) 2021. [DOI: 10.1002/pep2.24224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
14
|
Caporale A, Adorinni S, Lamba D, Saviano M. Peptide-Protein Interactions: From Drug Design to Supramolecular Biomaterials. Molecules 2021; 26:1219. [PMID: 33668767 PMCID: PMC7956380 DOI: 10.3390/molecules26051219] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The self-recognition and self-assembly of biomolecules are spontaneous processes that occur in Nature and allow the formation of ordered structures, at the nanoscale or even at the macroscale, under thermodynamic and kinetic equilibrium as a consequence of specific and local interactions. In particular, peptides and peptidomimetics play an elected role, as they may allow a rational approach to elucidate biological mechanisms to develop new drugs, biomaterials, catalysts, or semiconductors. The forces that rule self-recognition and self-assembly processes are weak interactions, such as hydrogen bonding, electrostatic attractions, and van der Waals forces, and they underlie the formation of the secondary structure (e.g., α-helix, β-sheet, polyproline II helix), which plays a key role in all biological processes. Here, we present recent and significant examples whereby design was successfully applied to attain the desired structural motifs toward function. These studies are important to understand the main interactions ruling the biological processes and the onset of many pathologies. The types of secondary structure adopted by peptides during self-assembly have a fundamental importance not only on the type of nano- or macro-structure formed but also on the properties of biomaterials, such as the types of interaction, encapsulation, non-covalent interaction, or covalent interaction, which are ultimately useful for applications in drug delivery.
Collapse
Affiliation(s)
- Andrea Caporale
- IC-CNR, c/o Area Science Park, S.S. 14 Km 163.5 Basovizza, 34149 Trieste, Italy;
| | - Simone Adorinni
- Dipartimento di Scienze Chimiche e Farmaceutiche di Università di Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy;
| | - Doriano Lamba
- IC-CNR, c/o Area Science Park, S.S. 14 Km 163.5 Basovizza, 34149 Trieste, Italy;
- Istituto Nazionale Biostrutture e Biosistemi, Consorzio Interuniversitario, Viale delle Medaglie d’Oro 305, I-00136 Roma, Italy
| | - Michele Saviano
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche (IC-CNR), Via Giovanni Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
15
|
Gelain F, Luo Z, Rioult M, Zhang S. Self-assembling peptide scaffolds in the clinic. NPJ Regen Med 2021; 6:9. [PMID: 33597509 PMCID: PMC7889856 DOI: 10.1038/s41536-020-00116-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Well-defined scaffold hydrogels made of self-assembling peptides have found their way into clinical products. By examining the properties and applications of two self-assembling peptides-EAK16 and RADA16-we highlight the potential for translating designer biological scaffolds into commercial products.
Collapse
Affiliation(s)
- Fabrizio Gelain
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBREMIT), IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy.
- Center for Nanomedicine and Tissue Engineering (CNTE), ASST Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy.
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Centre, Chongqing Medical University, Chongqing, 400016, China.
| | | | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, Cambridge, MA, 02139-4307, USA.
| |
Collapse
|
16
|
Morad T, Hendler RM, Canji E, Weiss OE, Sion G, Minnes R, Polaq AHG, Merfeld I, Dubinsky Z, Nesher E, Baranes D. Aragonite-Polylysine: Neuro-Regenerative Scaffolds with Diverse Effects on Astrogliosis. Polymers (Basel) 2020; 12:E2850. [PMID: 33260420 PMCID: PMC7760860 DOI: 10.3390/polym12122850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 02/03/2023] Open
Abstract
Biomaterials, especially when coated with adhesive polymers, are a key tool for restorative medicine, being biocompatible and supportive for cell adherence, growth, and function. Aragonite skeletons of corals are biomaterials that support survival and growth of a range of cell types, including neurons and glia. However, it is not known if this scaffold affects neural cell migration or elongation of neuronal and astrocytic processes, prerequisites for initiating repair of damage in the nervous system. To address this, hippocampal cells were aggregated into neurospheres and cultivated on aragonite skeleton of the coral Trachyphyllia geoffroyi (Coral Skeleton (CS)), on naturally occurring aragonite (Geological Aragonite (GA)), and on glass, all pre-coated with the oligomer poly-D-lysine (PDL). The two aragonite matrices promoted equally strong cell migration (4.8 and 4.3-fold above glass-PDL, respectively) and axonal sprouting (1.96 and 1.95-fold above glass-PDL, respectively). However, CS-PDL had a stronger effect than GA-PDL on the promotion of astrocytic processes elongation (1.7 vs. 1.2-fold above glass-PDL, respectively) and expression of the glial fibrillary acidic protein (3.8 vs. and 1.8-fold above glass-PDL, respectively). These differences are likely to emerge from a reaction of astrocytes to the degree of roughness of the surface of the scaffold, which is higher on CS than on GA. Hence, CS-PDL and GA-PDL are scaffolds of strong capacity to derive neural cell movements and growth required for regeneration, while controlling the extent of astrocytic involvement. As such, implants of PDL-aragonites have significant potential as tools for damage repair and the reduction of scar formation in the brain following trauma or disease.
Collapse
Affiliation(s)
- Tzachy Morad
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| | - Roni Mina Hendler
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| | - Eyal Canji
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| | - Orly Eva Weiss
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| | - Guy Sion
- School of Zoology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel;
- Institute for Land Water and Society, Charles Sturt University, P.O. Box 789, Elizabeth Mitchell Drive, Albury, NSW 2642, Australia
| | - Refael Minnes
- Department of Physics, Ariel University, Ariel 4070000, Israel;
| | - Ania Hava Grushchenko Polaq
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| | - Ido Merfeld
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| | - Zvy Dubinsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel;
| | - Elimelech Nesher
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
- Institute for Personalized and Translational Medicine, Ariel University, Ariel 4070000, Israel
| | - Danny Baranes
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; (T.M.); (R.M.H.); (E.C.); (O.E.W.); (A.H.G.P.); (I.M.); (E.N.)
| |
Collapse
|
17
|
Gelain F, Luo Z, Zhang S. Self-Assembling Peptide EAK16 and RADA16 Nanofiber Scaffold Hydrogel. Chem Rev 2020; 120:13434-13460. [DOI: 10.1021/acs.chemrev.0c00690] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Fabrizio Gelain
- Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013, Italy
- Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Piazza dell’Ospedale Maggiore, 3, Milan 20162, Italy
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139-4307, United States
| |
Collapse
|
18
|
Ding X, Zhao H, Li Y, Lee AL, Li Z, Fu M, Li C, Yang YY, Yuan P. Synthetic peptide hydrogels as 3D scaffolds for tissue engineering. Adv Drug Deliv Rev 2020; 160:78-104. [PMID: 33091503 DOI: 10.1016/j.addr.2020.10.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The regeneration of tissues and organs poses an immense challenge due to the extreme complexity in the research work involved. Despite the tissue engineering approach being considered as a promising strategy for more than two decades, a key issue impeding its progress is the lack of ideal scaffold materials. Nature-inspired synthetic peptide hydrogels are inherently biocompatible, and its high resemblance to extracellular matrix makes peptide hydrogels suitable 3D scaffold materials. This review covers the important aspects of peptide hydrogels as 3D scaffolds, including mechanical properties, biodegradability and bioactivity, and the current approaches in creating matrices with optimized features. Many of these scaffolds contain peptide sequences that are widely reported for tissue repair and regeneration and these peptide sequences will also be discussed. Furthermore, 3D biofabrication strategies of synthetic peptide hydrogels and the recent advances of peptide hydrogels in tissue engineering will also be described to reflect the current trend in the field. In the final section, we will present the future outlook in the design and development of peptide-based hydrogels for translational tissue engineering applications.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Ashlynn Lingzhi Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zongshao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengjing Fu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
19
|
Zhai H, Zhou J, Xu J, Sun X, Xu Y, Qiu X, Zhang C, Wu Z, Long H, Bai Y, Quan D. Mechanically strengthened hybrid peptide-polyester hydrogel and potential applications in spinal cord injury repair. Biomed Mater 2020; 15:055031. [DOI: 10.1088/1748-605x/ab9e45] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Mu X, Shi L, Pan S, He L, Niu Y, Wang X. A Customized Self-Assembling Peptide Hydrogel-Wrapped Stem Cell Factor Targeting Pulp Regeneration Rich in Vascular-Like Structures. ACS OMEGA 2020; 5:16568-16574. [PMID: 32685822 PMCID: PMC7364552 DOI: 10.1021/acsomega.0c01266] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/15/2020] [Indexed: 05/27/2023]
Abstract
Pulp regeneration is to replace the inflamed/necrotic pulp tissue with regenerated pulp-like tissue to rejuvenate the teeth. Self-assembling peptide hydrogels RADA16-I (Ac-(RADA16-I)4-CONH2) can provide a three-dimensional environment for cells. The stem cell factor (SCF) plays a crucial role in homing stem cells. Combining these advantages, our study investigated the effects of SCF-RADA16-I on adhesion, proliferation, and migration of human dental pulp stem cells (DPSCs) and the angiogenesis of human umbilical vein endothelial cells (HUVECs). The β-sheet and grid structure were observed by circular dichroism (CD), scanning electron microscopy (SEM), and atomic force microscopy (AFM). Cytoskeleton staining, living cell staining, cell viability, cell migration, angiogenesis, and western blot assays were performed, and the results indicated that all the SCF groups were superior to the corresponding non-SCF groups in cell adhesion, proliferation, migration, and angiogenesis. RADA16-I provided a three-dimensional environment for DPSCs. Besides, the SCF promoted HUVECs to form more vascular-like structures and release more vascular endothelial growth factor A. In summary, the SCF-loaded RADA16-I scaffold improved adhesion, proliferation, and migration of DPSCs and the formation of more vascular-like structures of HUVECs. SCF-RADA16-I holds promise for guided pulp regeneration, and it can potentially be applied widely in tissue engineering and translational medicine in the future.
Collapse
Affiliation(s)
- Xiaodan Mu
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Lei Shi
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Shuang Pan
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Lina He
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Yumei Niu
- The
First Affiliated Hospital, Harbin Medical
University, 23 Post Street, Nangang District, Harbin, Heilongjiang 150001, China
- School
of Stomatology, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang 150001, China
| | - Xiumei Wang
- Department
of Materials Science and Engineering, State Key Laboratory of New
Ceramics and Fine Processing, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing 100084, China
| |
Collapse
|
21
|
Zhao X, Zhu Y, Laslett AL, Chan HF. Hepatic Differentiation of Stem Cells in 2D and 3D Biomaterial Systems. Bioengineering (Basel) 2020; 7:E47. [PMID: 32466173 PMCID: PMC7356247 DOI: 10.3390/bioengineering7020047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023] Open
Abstract
A critical shortage of donor livers for treating end-stage liver failure signifies the urgent need for alternative treatment options. Hepatocyte-like cells (HLC) derived from various stem cells represent a promising cell source for hepatocyte transplantation, liver tissue engineering, and development of a bioartificial liver assist device. At present, the protocols of hepatic differentiation of stem cells are optimized based on soluble chemical signals introduced in the culture medium and the HLC produced typically retain an immature phenotype. To promote further hepatic differentiation and maturation, biomaterials can be designed to recapitulate cell-extracellular matrix (ECM) interactions in both 2D and 3D configurations. In this review, we will summarize and compare various 2D and 3D biomaterial systems that have been applied to hepatic differentiation, and highlight their roles in presenting biochemical and physical cues to different stem cell sources.
Collapse
Affiliation(s)
- Xiaoyu Zhao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yanlun Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Andrew L. Laslett
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia;
- Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; (X.Z.); (Y.Z.)
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
22
|
Glotzbach K, Stamm N, Weberskirch R, Faissner A. Hydrogels Derivatized With Cationic Moieties or Functional Peptides as Efficient Supports for Neural Stem Cells. Front Neurosci 2020; 14:475. [PMID: 32508574 PMCID: PMC7251306 DOI: 10.3389/fnins.2020.00475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
The increasing incidence of neurodegenerative diseases such as Alzheimer's or Parkinson's disease represents a significant burden for patients and national health systems. The conditions are primarily caused by the death of neurons and other neural cell types. One important aim of current stem cell research is to find a way to replace the lost cells. In this perspective, neural stem cells (NSCs) have been considered as a promising tool in the field of regenerative medicine. The behavior of NSCs is modulated by environmental influences, for example hormones, growth factors, cytokines, and extracellular matrix molecules or biomechanics. These factors can be studied by using well-defined hydrogels, which are polymeric networks of synthetic or natural origin with the ability to swell in water. These gels can be modified with a variety of molecules and optimized with regard to their mechanical properties to mimic the natural extracellular environment. In particular modifications applying distinct units such as functional domains and peptides can modulate the development of NSCs with regard to proliferation, differentiation and migration. One well-known peptide sequence that affects the behavior of NSCs is the integrin recognition sequence RGD that has originally been derived from fibronectin. In the present review we provide an overview concerning the applications of modified hydrogels with an emphasis on synthetic hydrogels based on poly(acrylamides), as modified with either cationic moieties or the peptide sequence RGD. This knowledge might be used in tissue engineering and regenerative medicine for the therapy of spinal cord injuries, neurodegenerative diseases and traumata.
Collapse
Affiliation(s)
- Kristin Glotzbach
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Nils Stamm
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Ralf Weberskirch
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
23
|
Marchini A, Favoino C, Gelain F. Multi-Functionalized Self-Assembling Peptides as Reproducible 3D Cell Culture Systems Enabling Differentiation and Survival of Various Human Neural Stem Cell Lines. Front Neurosci 2020; 14:413. [PMID: 32431590 PMCID: PMC7214803 DOI: 10.3389/fnins.2020.00413] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Neural stem cells-based therapies have shown great potential for central nervous system regeneration, with three-dimensional (3D) culture systems representing a key technique for tissue engineering applications, as well as disease modeling and drug screenings. Self-assembling peptides (SAPs), providing biomimetic synthetic micro-environments regulating cellular functionality and tissue repair, constitute a suitable tool for the production of complex tissue-like structures in vitro. However, one of the most important drawbacks in 3D cultures, obtained via animal-derived substrates and serum-rich media, is the reproducibility and tunability of a standardized methodology capable to coax neural differentiation of different human cell lines. In this work we cultured four distinct human neural stem cell (hNSC) lines in 3D synthetic multifunctionalized hydrogel (named HYDROSAP) for up to 6 weeks. Three-dimensional cultures of differentiating hNSCs exhibited a progressive differentiation and maturation over time. All hNSCs-derived neurons in 3D culture system exhibited randomly organized entangled networks with increasing expression of GABAergic and glutamatergic phenotypes and presence of cholinergic ones. Oligodendrocytes formed insulating myelin sheaths positive for myelin basic protein (MBP). In summary, results demonstrated a successfully standardized and reproducible 3D cell culture system for hNSC differentiation and maturation in serum-free conditions useful for future therapies.
Collapse
Affiliation(s)
- Amanda Marchini
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Chiara Favoino
- Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Fabrizio Gelain
- Tissue Engineering Unit, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies-ISBReMIT, Fondazione IRCSS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Center for Nanomedicine and Tissue Engineering, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
24
|
Bordoni M, Scarian E, Rey F, Gagliardi S, Carelli S, Pansarasa O, Cereda C. Biomaterials in Neurodegenerative Disorders: A Promising Therapeutic Approach. Int J Mol Sci 2020; 21:ijms21093243. [PMID: 32375302 PMCID: PMC7247337 DOI: 10.3390/ijms21093243] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and spinal cord injury) represent a great problem worldwide and are becoming prevalent because of the increasing average age of the population. Despite many studies having focused on their etiopathology, the exact cause of these diseases is still unknown and until now, there are only symptomatic treatments. Biomaterials have become important not only for the study of disease pathogenesis, but also for their application in regenerative medicine. The great advantages provided by biomaterials are their ability to mimic the environment of the extracellular matrix and to allow the growth of different types of cells. Biomaterials can be used as supporting material for cell proliferation to be transplanted and as vectors to deliver many active molecules for the treatments of neurodegenerative disorders. In this review, we aim to report the potentiality of biomaterials (i.e., hydrogels, nanoparticles, self-assembling peptides, nanofibers and carbon-based nanomaterials) by analyzing their use in the regeneration of neural and glial cells their role in axon outgrowth. Although further studies are needed for their use in humans, the promising results obtained by several groups leads us to suppose that biomaterials represent a potential therapeutic approach for the treatments of neurodegenerative disorders.
Collapse
Affiliation(s)
- Matteo Bordoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy;
| | - Eveljn Scarian
- Department of Brain and Behavioural Sciences, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy;
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
| | - Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (S.C.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milan, Via Grassi, 74, 20157 Milan, Italy
| | - Stella Gagliardi
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (S.C.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milan, Via Grassi, 74, 20157 Milan, Italy
| | - Orietta Pansarasa
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
- Correspondence: ; Tel.: +39-0382-380-248
| | - Cristina Cereda
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (S.G.); (C.C.)
| |
Collapse
|
25
|
Revkova VA, Grebenik EA, Kalsin VA, Demina TS, Bardakova KN, Shavkuta BS, Melnikov PA, Samoilova EM, Konoplyannikov MA, Efremov YM, Zhang C, Akopova TA, Troitsky AV, Timashev PS, Baklaushev VP. Chitosan- g-oligo(L,L-lactide) Copolymer Hydrogel Potential for Neural Stem Cell Differentiation. Tissue Eng Part A 2020; 26:953-963. [PMID: 32159465 DOI: 10.1089/ten.tea.2019.0265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We evaluated the applicability of chitosan-g-oligo(L,L-lactide) copolymer (CLC) hydrogel for central nervous system tissue engineering. The biomechanical properties of the CLC hydrogel were characterized and its biocompatibility was assessed with neural progenitor cells obtained from two different sources: H9-derived neural stem cells (H9D-NSCs) and directly reprogrammed neural precursor cells (drNPCs). Our study found that the optically transparent CLC hydrogel possessed biomechanical characteristics suitable for culturing human neural stem/precursor cells and was noncytotoxic. When seeded on films prepared from CLC copolymer hydrogel, both H9D-NSC and drNPC adhered well, expanded and exhibited signs of spontaneous differentiation. While H9D-NSC mainly preserved multipotency as shown by a high proportion of Nestin+ and Sox2+ cells and a comparatively lower expression of the neuronal markers βIII-tubulin and MAP2, drNPCs, obtained by direct reprogramming, differentiated more extensively along the neuronal lineage. Our study indicates that the CLC hydrogel may be considered as a substrate for tissue-engineered constructs, applicable for therapy of neurodegenerative diseases. Impact statement We synthetized a chitosan-g-oligo(L,L-lactide) hydrogel that sustained multipotency of embryonic-derived neural stem cells (NSCs) and supported differentiation of directly reprogrammed NSC predominantly along the neuronal lineage. The hydrogel exhibited no cytotoxicity in vitro, both in extraction and contact cytotoxicity tests. When seeded on the hydrogel, both types of NSCs adhered well, expanded, and exhibited signs of spontaneous differentiation. The biomechanical properties of the hydrogel were similar to that of human spinal cord with incised pia mater. These data pave the way for further investigations of the hydrogel toward its applicability in central nervous system tissue engineering.
Collapse
Affiliation(s)
- Veronica A Revkova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Ekaterina A Grebenik
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir A Kalsin
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Tatiana S Demina
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Enikolopov Institute of Synthetic Polymer Materials, Russian Academy of Sciences, Moscow, Russia
| | - Kseniia N Bardakova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Research Center "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Boris S Shavkuta
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Research Center "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia
| | - Pavel A Melnikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia.,Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Ekaterina M Samoilova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Mikhail A Konoplyannikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia.,Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yuri M Efremov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumors, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tatiana A Akopova
- Enikolopov Institute of Synthetic Polymer Materials, Russian Academy of Sciences, Moscow, Russia
| | - Alexandr V Troitsky
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| | - Peter S Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Institute of Photonic Technologies, Research Center "Crystallography and Photonics," Russian Academy of Sciences, Moscow, Russia.,N.N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir P Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia
| |
Collapse
|
26
|
Li H, Chen S, Piao S, An T, Wang C. Production of artificial synthetic spidroin gene 4S-transgenic cloned sheep embryos using somatic cell nuclear transfer. Anim Biotechnol 2020; 32:616-626. [DOI: 10.1080/10495398.2020.1737098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Hao Li
- College of Life Sciences, Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
- College of Life Science, Laboratory of Developmental Biology, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
| | - Shengnan Chen
- College of Life Sciences, Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
| | - Shanhua Piao
- College of Life Sciences, Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
| | - Tiezhu An
- College of Life Sciences, Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
- College of Life Science, Laboratory of Developmental Biology, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
| | - Chunsheng Wang
- College of Life Sciences, Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
- College of Life Science, Laboratory of Developmental Biology, Northeast Forestry University, Harbin, Heilongjiang, P. R. China
| |
Collapse
|
27
|
Huang H, Zhong L, Zhou J, Hou Y, Zhang Z, Xing X, Sun J. Leydig-like cells derived from reprogrammed human foreskin fibroblasts by CRISPR/dCas9 increase the level of serum testosterone in castrated male rats. J Cell Mol Med 2020; 24:3971-3981. [PMID: 32160419 PMCID: PMC7171312 DOI: 10.1111/jcmm.15018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/28/2019] [Accepted: 01/06/2020] [Indexed: 12/13/2022] Open
Abstract
In the past few years, Leydig cell (LC) transplantation has been regarded as an effective strategy for providing physiological patterns of testosterone in vivo. Recently, we have successfully converted human foreskin fibroblasts (HFFs) into functional Leydig‐like cells (iLCs) in vitro by using the CRISPR/dCas9 system, which shows promising potential for seed cells. However, it is not known whether the reprogrammed iLCs can survive or restore serum testosterone levels in vivo. Therefore, in this study, we evaluate whether reprogrammed iLCs can restore the serum testosterone levels of castrated rats when they are transplanted into the fibrous capsule. We first developed the castrated Sprague Dawley rat model through bilateral orchiectomy and subsequently injected extracellular matrix gel containing transplanted cells into the fibrous capsule of castrated rats. Finally, we evaluated dynamic serum levels of testosterone and luteinizing hormone (LH) in castrated rats, the survival of implanted iLCs, and the expression levels of Leydig steroidogenic enzymes by immunofluorescence staining and Western blotting. Our results demonstrated that implanted iLCs could partially restore the serum testosterone level of castrated rats, weakly mimic the role of adult Leydig cells in the hypothalamic‐pituitary‐gonadal axis for a short period, and survive and secrete testosterone, through 6 weeks after transplantation. Therefore, this study may be valuable for treating male hypogonadism in the future.
Collapse
Affiliation(s)
- Hua Huang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhong
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Zhou
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanping Hou
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Zhang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Xing
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Ghane N, Beigi MH, Labbaf S, Nasr-Esfahani MH, Kiani A. Design of hydrogel-based scaffolds for the treatment of spinal cord injuries. J Mater Chem B 2020; 8:10712-10738. [DOI: 10.1039/d0tb01842b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hydrogel-based scaffold design approaches for the treatment of spinal cord injuries.
Collapse
Affiliation(s)
- Nazanin Ghane
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Mohammad-Hossein Beigi
- Department of Cellular Biotechnology Cell Science Research Center
- Royan Institute for Biotechnology
- ACECR
- Isfahan
- Iran
| | - Sheyda Labbaf
- Biomaterials Research Group
- Department of Materials Engineering
- Isfahan University of Technology
- Isfahan
- Iran
| | | | - Amirkianoosh Kiani
- Silicon Hall: Micro/Nano Manufacturing Facility
- Faculty of Engineering and Applied Science
- Ontario Tech University
- Ontario
- Canada
| |
Collapse
|
29
|
Hong YJ, Do JT. Neural Lineage Differentiation From Pluripotent Stem Cells to Mimic Human Brain Tissues. Front Bioeng Biotechnol 2019; 7:400. [PMID: 31867324 PMCID: PMC6908493 DOI: 10.3389/fbioe.2019.00400] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/22/2019] [Indexed: 01/22/2023] Open
Abstract
Recent advances in induced pluripotent stem cell (iPSC) research have turned limitations of prior and current research into possibilities. iPSCs can differentiate into the desired cell types, are easier to obtain than embryonic stem cells (ESCs), and more importantly, in case they are to be used in research on diseases, they can be obtained directly from the patient. With these advantages, differentiation of iPSCs into various cell types has been conducted in the fields of basic development, cell physiology, and cell therapy research. Differentiation of stem cells into nervous cells has been prevalent among all cell types studied. Starting with the monolayer 2D differentiation method where cells were attached to a dish, substantial efforts have been made to better mimic the in vivo environment and produce cells grown in vitro that closely resemble in vivo state cells. Having surpassed the stage of 3D differentiation, we have now reached the stage of creating tissues called organoids that resemble organs, rather than growing simple cells. In this review, we focus on the central nervous system (CNS) and describe the challenges faced in 2D and 3D differentiation research studies and the processes of overcoming them. We also discuss current studies and future perspectives on brain organoid researches.
Collapse
Affiliation(s)
- Yean Ju Hong
- Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology, Konkuk University, Seoul, South Korea
| | - Jeong Tae Do
- Department of Stem Cell and Regenerative Biotechnology, KU Institute of Science and Technology, Konkuk University, Seoul, South Korea
| |
Collapse
|
30
|
Lee S, Trinh TH, Yoo M, Shin J, Lee H, Kim J, Hwang E, Lim YB, Ryou C. Self-Assembling Peptides and Their Application in the Treatment of Diseases. Int J Mol Sci 2019; 20:E5850. [PMID: 31766475 PMCID: PMC6928719 DOI: 10.3390/ijms20235850] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
Self-assembling peptides are biomedical materials with unique structures that are formed in response to various environmental conditions. Governed by their physicochemical characteristics, the peptides can form a variety of structures with greater reactivity than conventional non-biological materials. The structural divergence of self-assembling peptides allows for various functional possibilities; when assembled, they can be used as scaffolds for cell and tissue regeneration, and vehicles for drug delivery, conferring controlled release, stability, and targeting, and avoiding side effects of drugs. These peptides can also be used as drugs themselves. In this review, we describe the basic structure and characteristics of self-assembling peptides and the various factors that affect the formation of peptide-based structures. We also summarize the applications of self-assembling peptides in the treatment of various diseases, including cancer. Furthermore, the in-cell self-assembly of peptides, termed reverse self-assembly, is discussed as a novel paradigm for self-assembling peptide-based nanovehicles and nanomedicines.
Collapse
Affiliation(s)
- Sungeun Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Trang H.T. Trinh
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Miryeong Yoo
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Junwu Shin
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Hakmin Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Jaehyeon Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| | - Euimin Hwang
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Korea; (E.H.); (Y.-b.L.)
| | - Yong-beom Lim
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Korea; (E.H.); (Y.-b.L.)
| | - Chongsuk Ryou
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Gyeonggi-do 15588, Korea; (S.L.); (M.Y.); (J.S.); (H.L.); (J.K.)
| |
Collapse
|
31
|
Sharma P, Kaur H, Roy S. Designing a Tenascin-C-Inspired Short Bioactive Peptide Scaffold to Direct and Control Cellular Behavior. ACS Biomater Sci Eng 2019; 5:6497-6510. [DOI: 10.1021/acsbiomaterials.9b01115] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Pooja Sharma
- Institute of Nanoscience and Technology, Habitat Centre, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Harsimran Kaur
- Institute of Nanoscience and Technology, Habitat Centre, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Sangita Roy
- Institute of Nanoscience and Technology, Habitat Centre, Sector 64, Phase 10, Mohali, Punjab 160062, India
| |
Collapse
|
32
|
Unal DB, Caliari SR, Lampe KJ. Engineering biomaterial microenvironments to promote myelination in the central nervous system. Brain Res Bull 2019; 152:159-174. [PMID: 31306690 DOI: 10.1016/j.brainresbull.2019.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023]
Abstract
Promoting remyelination and/or minimizing demyelination are key therapeutic strategies under investigation for diseases and injuries like multiple sclerosis (MS), spinal cord injury, stroke, and virus-induced encephalopathy. Myelination is essential for efficacious neuronal signaling. This myelination process is originated by oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). Resident OPCs are capable of both proliferation and differentiation, and also migration to demyelinated injury sites. OPCs can then engage with these unmyelinated or demyelinated axons and differentiate into myelin-forming oligodendrocytes (OLs). However this process is frequently incomplete and often does not occur at all. Biomaterial strategies can now be used to guide OPC and OL development with the goal of regenerating healthy myelin sheaths in formerly damaged CNS tissue. Growth and neurotrophic factors delivered from such materials can promote proliferation of OPCs or differentiation into OLs. While cell transplantation techniques have been used to replace damaged cells in wound sites, they have also resulted in poor transplant cell viability, uncontrollable differentiation, and poor integration into the host. Biomaterial scaffolds made from extracellular matrix (ECM) mimics that are naturally or synthetically derived can improve transplanted cell survival, support both transplanted and endogenous cell populations, and direct their fate. In particular, stiffness and degradability of these scaffolds are two parameters that can influence the fate of OPCs and OLs. The future outlook for biomaterials research includes 3D in vitro models of myelination / remyelination / demyelination to better mimic and study these processes. These models should provide simple relationships of myelination to microenvironmental biophysical and biochemical properties to inform improved therapeutic approaches.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States.
| |
Collapse
|
33
|
Arora N, Shome R, Ghosh SS. Deciphering therapeutic potential of PEGylated recombinant PTEN-silver nanoclusters ensemble on 3D spheroids. Mol Biol Rep 2019; 46:5103-5112. [PMID: 31290055 DOI: 10.1007/s11033-019-04965-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
The therapeutic application of recombinant proteins is limited due to their inherent structural complexity. Additionally, screening of therapeutic potential of protein products requires an appropriate testing platform to achieve biological relevance. Fabrication of three dimensional cultures bridges the gap between in vitro based monolayer cultures and clinical applications. In this perspective, glioblastoma U-87 MG and breast cancer MCF7 spheroids were generated to assess the therapeutic prospect of recombinant PTEN protein. PTEN bound to silver nanoclusters was encapsulated within PEG coating, which resulted in fabrication of spherical nanocarriers named as PTEN-nanocomposites. Internalization of PTEN-nanocomposites in the spheroids was confirmed by confocal microscopy. Upon uptake, PTEN-nanocomposites led to modulation of cyclins and apoptosis gene regulators culminating in cell cycle arrest and reduced cell viability as confirmed by calcein-AM/PI dual staining and alamar blue assay. Further, combination of tamoxifen and PTEN-nanocomposites on U-87 MG spheroids resulted in two-fold reduction of drug dosage. The study revealed that the monolayer culture results translated to the 3D culture as well, however higher dose of the recombinant PTEN was required for the spheroid system. The anti-proliferative role of PTEN-nanocomposites in a complex 3D environment augments its biological implication and paves the way for recombinant PTEN based therapeutic applications.
Collapse
Affiliation(s)
- Neha Arora
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Rajib Shome
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India. .,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
34
|
Nune M, Subramanian A, Krishnan UM, Sethuraman S. Peptide nanostructures on nanofibers for peripheral nerve regeneration. J Tissue Eng Regen Med 2019; 13:1059-1070. [PMID: 30946535 DOI: 10.1002/term.2860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 12/06/2018] [Accepted: 02/13/2019] [Indexed: 01/04/2023]
Abstract
Self-assembled peptide nanofibrous scaffolds with designer sequences, similar to neurite growth promoting molecules enhance the differentiation of neural stem cells. However, self-assembled peptide nanofibrous scaffolds lack the required mechanical strength to suffice to bridge long critical-sized peripheral nerve defects. Hence, there is a demand for a potential neural substrate, which could be biomimetic coupled with bioactive nanostructures to regrow the denuded axons towards the distal end. In the present study, we developed designer self-assembling peptide-based aligned poly(lactic-co-glycolic acid) (PLGA) nanofibrous scaffolds by simple surface coating of peptides or coelectrospinning. Retention of secondary structures of peptides in peptide-coated and cospun fibers was confirmed by circular dichroism spectroscopy. The rod-like peptide nanostructures enhance the typical bipolar morphology of Schwann cells. Although the peptide-coated PLGA scaffolds exhibited significant increase in Schwann cell proliferation than pristine PLGA and PLGA-peptide cospun scaffolds (p < .05), peptide cospun scaffolds demonstrated better cellular infiltration and significantly higher gene expression of neural cell adhesion molecule, glial fibrillary acidic protein, and peripheral myelin protein22 compared to the pristine PLGA and PLGA-peptide-coated scaffolds. Our results demonstrate the positive effects of aligned peptide coelectrospun scaffolds with biomimetic cell recognition motifs towards functional proliferation of Schwann cells. These scaffolds could subsequently repair peripheral nerve defects by augmenting axonal regeneration and functional nerve recovery.
Collapse
Affiliation(s)
- Manasa Nune
- Centre of Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Anuradha Subramanian
- Centre of Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Uma Maheswari Krishnan
- Centre of Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Swaminathan Sethuraman
- Centre of Nanotechnology & Advanced Biomaterials, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
35
|
Determination of mechanical and rheological properties of a cell-loaded peptide gel during ECM production. Int J Pharm 2019; 563:437-444. [DOI: 10.1016/j.ijpharm.2019.04.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022]
|
36
|
Cao B, Li Y, Yang T, Bao Q, Yang M, Mao C. Bacteriophage-based biomaterials for tissue regeneration. Adv Drug Deliv Rev 2019; 145:73-95. [PMID: 30452949 PMCID: PMC6522342 DOI: 10.1016/j.addr.2018.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 07/24/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
Abstract
Bacteriophage, also called phage, is a human-safe bacteria-specific virus. It is a monodisperse biological nanostructure made of proteins (forming the outside surface) and nucleic acids (encased in the protein capsid). Among different types of phages, filamentous phages have received great attention in tissue regeneration research due to their unique nanofiber-like morphology. They can be produced in an error-free format, self-assemble into ordered scaffolds, display multiple signaling peptides site-specifically, and serve as a platform for identifying novel signaling or homing peptides. They can direct stem cell differentiation into specific cell types when they are organized into proper patterns or display suitable peptides. These unusual features have allowed scientists to employ them to regenerate a variety of tissues, including bone, nerves, cartilage, skin, and heart. This review will summarize the progress in the field of phage-based tissue regeneration and the future directions in this field.
Collapse
Affiliation(s)
- Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Qing Bao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Zhejiang, Hangzhou 310058, China.
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States; School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China.
| |
Collapse
|
37
|
Pugliese R, Maleki M, Zuckermann RN, Gelain F. Self-assembling peptides cross-linked with genipin: resilient hydrogels and self-standing electrospun scaffolds for tissue engineering applications. Biomater Sci 2019; 7:76-91. [PMID: 30475373 DOI: 10.1039/c8bm00825f] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Self-assembling peptides (SAPs) are synthetic bioinspired biomaterials that can be feasibly multi-functionalized for applications in surgery, drug delivery, optics and tissue engineering (TE). Despite their promising biocompatibility and biomimetic properties, they have never been considered real competitors of polymers and/or cross-linked extracellular matrix (ECM) natural proteins. Indeed, synthetic SAP-made hydrogels usually feature modest mechanical properties, limiting their potential applications, due to the transient non-covalent interactions involved in the self-assembling phenomenon. Cross-linked SAP-hydrogels have been recently introduced to bridge this gap, but several questions remain open. New strategies leading to stiffer gels of SAPs may allow for a full exploitation of the SAP technology in TE and beyond. We have developed and characterized a genipin cross-linking strategy significantly increasing the stiffness and resiliency of FAQ(LDLK)3, a functionalized SAP already used for nervous cell cultures. We characterized different protocols of cross-linking, analyzing their dose and time-dependent efficiency, influencing stiffness, bioabsorption time and molecular arrangements. We choose the best developed protocol to electrospin into nanofibers, for the first time, self-standing, water-stable and flexible fibrous mats and micro-channels entirely made of SAPs. This work may open the door to the development and tailoring of bioprostheses entirely made of SAPs for different TE applications.
Collapse
Affiliation(s)
- Raffaele Pugliese
- IRCSS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, FG 71013, Italy.
| | | | | | | |
Collapse
|
38
|
Multifunctionalized hydrogels foster hNSC maturation in 3D cultures and neural regeneration in spinal cord injuries. Proc Natl Acad Sci U S A 2019; 116:7483-7492. [PMID: 30923117 PMCID: PMC6462084 DOI: 10.1073/pnas.1818392116] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cells reside in 3D microenvironments in living tissues; consequently, 3D cultures gained great interest because they better mimic the natural conditions of cells. Self-assembling peptides (SAPs) are synthetic bioabsorbable biomaterials that can provide customized 3D microenvironments regulating cell functionalities and tissue repair. Here we introduce a SAP-hydrogel designed to support human neural stem cell (hNSC) differentiation in 3D serum-free conditions, generating mature and active human neurons in vitro. We also demonstrate its functional neurorigenerative potential in rat spinal cord injuries, peaking when seeded with hNSCs progeny predifferentiated in vitro for 6 weeks. Despite these promising results, this approach should be confirmed in the future with medium-size animal models and with additional and refined behavioral tests before entering a clinical trial. Three-dimensional cell cultures are leading the way to the fabrication of tissue-like constructs useful to developmental biology and pharmaceutical screenings. However, their reproducibility and translational potential have been limited by biomaterial and culture media compositions, as well as cellular sources. We developed a construct comprising synthetic multifunctionalized hydrogels, serum-free media, and densely seeded good manufacturing practice protocol-grade human neural stem cells (hNSC). We tracked hNSC proliferation, differentiation, and maturation into GABAergic, glutamatergic, and cholinergic neurons, showing entangled electrically active neural networks. The neuroregenerative potential of the “engineered tissue” was assessed in spinal cord injuries, where hNSC-derived progenitors and predifferentiated hNSC progeny, embedded in multifunctionalized hydrogels, were implanted. All implants decreased astrogliosis and lowered the immune response, but scaffolds with predifferentiated hNSCs showed higher percentages of neuronal markers, better hNSC engraftment, and improved behavioral recovery. Our hNSC-construct enables the formation of 3D functional neuronal networks in vitro, allowing novel strategies for hNSC therapies in vivo.
Collapse
|
39
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
40
|
Cellular Spheroids of Mesenchymal Stem Cells and Their Perspectives in Future Healthcare. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9040627] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intrinsic cellular properties of several types of cells are dramatically altered as the culture condition shifts from two-dimensional (2D) to three-dimensional (3D) environment. Currently, several lines of evidence have demonstrated the therapeutic potential of mesenchymal stem cells (MSCs) in regenerative medicine. MSCs not only replenish the lost cells, they also promote the regeneration of impaired tissues by modulating the immune responses. Following the development of 3D cell culture, the enhanced therapeutic efficacy of spheroid-forming MSCs have been identified in several animal disease models by promoting differentiation or trophic factor secretion, as compared to planar-cultured MSCs. Due to the complicated and multifunctional applications in the medical field, MSCs are recently named as medicinal signaling cells. In this review, we summarize the predominant differences of cell–environment interactions for the MSC spheroids formed by chitosan-based substrates and other scaffold-free approaches. Furthermore, several important physical and chemical factors affecting cell behaviors in the cell spheroids are discussed. Currently, the understanding of MSCs spheroid interactions is continuously expanding. Overall, this article aims to review the broad advantages and perspectives of MSC spheroids in regenerative medicine and in future healthcare.
Collapse
|
41
|
Kornev VA, Grebenik EA, Solovieva AB, Dmitriev RI, Timashev PS. Hydrogel-assisted neuroregeneration approaches towards brain injury therapy: A state-of-the-art review. Comput Struct Biotechnol J 2018; 16:488-502. [PMID: 30455858 PMCID: PMC6232648 DOI: 10.1016/j.csbj.2018.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022] Open
Abstract
Recent years have witnessed the development of an enormous variety of hydrogel-based systems for neuroregeneration. Formed from hydrophilic polymers and comprised of up to 90% of water, these three-dimensional networks are promising tools for brain tissue regeneration. They can assist structural and functional restoration of damaged tissues by providing mechanical support and navigating cell fate. Hydrogels also show the potential for brain injury therapy due to their broadly tunable physical, chemical, and biological properties. Hydrogel polymers, which have been extensively implemented in recent brain injury repair studies, include hyaluronic acid, collagen type I, alginate, chitosan, methylcellulose, Matrigel, fibrin, gellan gum, self-assembling peptides and proteins, poly(ethylene glycol), methacrylates, and methacrylamides. When viewed as tools for neuroregeneration, hydrogels can be divided into: (1) hydrogels suitable for brain injury therapy, (2) hydrogels that do not meet basic therapeutic requirements and (3) promising hydrogels which meet the criteria for further investigations. Our analysis shows that fibrin, collagen I and self-assembling peptide-based hydrogels display very attractive properties for neuroregeneration.
Collapse
Affiliation(s)
- Vladimir A. Kornev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Ekaterina A. Grebenik
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Anna B. Solovieva
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
| | - Ruslan I. Dmitriev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
- Institute of Photonic Technologies, Research Center “Crystallography and Photonics” Russian Academy of Sciences, 2 Pionerskaya st., Troitsk, Moscow 108840, Russian Federation
| |
Collapse
|
42
|
Koch F, Wolff A, Mathes S, Pieles U, Saxer SS, Kreikemeyer B, Peters K. Amino acid composition of nanofibrillar self-assembling peptide hydrogels affects responses of periodontal tissue cells in vitro. Int J Nanomedicine 2018; 13:6717-6733. [PMID: 30425485 PMCID: PMC6204879 DOI: 10.2147/ijn.s173702] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background The regeneration of tissue defects at the interface between soft and hard tissue, eg, in the periodontium, poses a challenge due to the divergent tissue requirements. A class of biomaterials that may support the regeneration at the soft-to-hard tissue interface are self-assembling peptides (SAPs), as their physicochemical and mechanical properties can be rationally designed to meet tissue requirements. Materials and methods In this work, we investigated the effect of two single-component and two complementary β-sheet forming SAP systems on their hydrogel properties such as nanofibrillar architecture, surface charge, and protein adsorption as well as their influence on cell adhesion, morphology, growth, and differentiation. Results We showed that these four 11-amino acid SAP (P11-SAP) hydrogels possessed physico-chemical characteristics dependent on their amino acid composition that allowed variabilities in nanofibrillar network architecture, surface charge, and protein adsorption (eg, the single-component systems demonstrated an ~30% higher porosity and an almost 2-fold higher protein adsorption compared with the complementary systems). Cytocompatibility studies revealed similar results for cells cultured on the four P11-SAP hydrogels compared with cells on standard cell culture surfaces. The single-component P11-SAP systems showed a 1.7-fold increase in cell adhesion and cellular growth compared with the complementary P11-SAP systems. Moreover, significantly enhanced osteogenic differentiation of human calvarial osteoblasts was detected for the single-component P11-SAP system hydrogels compared with standard cell cultures. Conclusion Thus, single-component system P11-SAP hydrogels can be assessed as suitable scaffolds for periodontal regeneration therapy, as they provide adjustable, extracellular matrix-mimetic nanofibrillar architecture and favorable cellular interaction with periodontal cells.
Collapse
Affiliation(s)
- Franziska Koch
- Institute for Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland.,Department of Cell Biology, University Medicine Rostock, Rostock, Germany, .,Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Anne Wolff
- Department of Cell Biology, University Medicine Rostock, Rostock, Germany,
| | - Stephanie Mathes
- Department for Chemistry and Biotechnology, Tissue Engineering, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Uwe Pieles
- Institute for Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Sina S Saxer
- Institute for Chemistry and Bioanalytics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Kirsten Peters
- Department of Cell Biology, University Medicine Rostock, Rostock, Germany,
| |
Collapse
|
43
|
Hainline KM, Gu F, Handley JF, Tian YF, Wu Y, de Wet L, Vander Griend DJ, Collier JH. Self-Assembling Peptide Gels for 3D Prostate Cancer Spheroid Culture. Macromol Biosci 2018; 19:e1800249. [PMID: 30324687 DOI: 10.1002/mabi.201800249] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Progress in prostate cancer research is presently limited by a shortage of reliable in vitro model systems. The authors describe a novel self-assembling peptide, bQ13, which forms nanofibers and gels useful for the 3D culture of prostate cancer spheroids, with improved cytocompatibility compared to related fibrillizing peptides. The mechanical properties of bQ13 gels can be controlled by adjusting peptide concentration, with storage moduli ranging between 1 and 10 kPa. bQ13's ability to remain soluble at mildly basic pH considerably improved the viability of encapsulated cells compared to other self-assembling nanofiber-forming peptides. LNCaP cells formed spheroids in bQ13 gels with similar morphologies and sizes to those formed in Matrigel or RADA16-I. Moreover, prostate-specific antigen (PSA) is produced by LNCaP cells in all matrices, and PSA production is more responsive to enzalutamide treatment in bQ13 gels than in other fibrillized peptide gels. bQ13 represents an attractive platform for further tailoring within 3D cell culture systems.
Collapse
Affiliation(s)
- Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Fangqi Gu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jacqueline F Handley
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Ye F Tian
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Larischa de Wet
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Donald J Vander Griend
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
44
|
Pugliese R, Fontana F, Marchini A, Gelain F. Branched peptides integrate into self-assembled nanostructures and enhance biomechanics of peptidic hydrogels. Acta Biomater 2018; 66:258-271. [PMID: 29128535 DOI: 10.1016/j.actbio.2017.11.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
Self-assembling peptides (SAP) have drawn an increasing interest in the tissue engineering community. They display unquestionable biomimetic properties, tailorability and promising biocompatibility. However their use has been hampered by poor mechanical properties making them fragile soft scaffolds. To increase SAP hydrogel stiffness we introduced a novel strategy based on multiple ramifications of (LDLK)3, a well-known linear SAP, connected with one or multiple "lysine knots". Differently branched SAPs were tested by increasing the number of (LDLK)3-like branches and by adding the neuro-regenerative functional motif BMHP1 as a single branch. While pure branched peptides did not have appealing self-assembling propensity, when mixed with the corresponding linear SAP they increased the stiffness of the overall hydrogel of multiple times. Notably, optimal results (or peak) were obtained 1) at similar molar ratio (between linear and branched peptides) for all tested sequences and 2) for the branched SAPs featuring the highest number of branches made of (LDLK)3. The functional motif BMHP1, as expected, seemed not to contribute to the increase of the storage modulus as efficiently as (LDLK)3. Interestingly, branched SAPs improved the β-sheet self-arrangement of (LDLK)3 and allowed for the formation of assembled nanofibers. Indeed in coarse-grained molecular dynamics we showed they readily integrate in the assembled aggregates providing "molecular connections" among otherwise weakly paired β-structures. Lastly, branched SAPs did not affect the usual response of human neural stem cells cultured on (LDLK)3-like scaffolds in vitro. Hence, branched SAPs may be a valuable new tool to enhance mechanical properties of self-assembling peptide biomaterials harmlessly; as neither chemical nor enzymatic cross-linking reactions are involved. As a consequence, branched SAPs may enlarge the field of application of SAPs in tissue engineering and beyond. STATEMENT OF SIGNIFICANCE Self-assembling peptides stand at the forefront of regenerative medicine because they feature biomimetic nano-architectures that mimic the complexity of natural peptide-based extracellular matrices of living tissues. Their superior biocompatibility and ease of scale-up production are hampered by weak mechanical properties due to transient non-covalent interactions among and within the self-assembled peptide chains, thus limiting their potential applications. We introduced new branched self-assembling peptides to be used as "molecular connectors" among self-assembled nanostructures made of linear SAPs. Branched SAPs could be mixed with linear SAPs before self-assembling in order to have them intermingled with different β-sheets of linear SAPs after gelation. This strategy caused a manifold increase of the stiffness of the assembled hydrogels (proportional to the number of self-assembling branches), did not affect SAP propensity to form β-sheet but, instead, further stimulated their secondary structure rearrangements. It is now possible to modularly improve SAP scaffold mechanical properties without using harmful chemical reactions. Therefore, branched SAPs represent an additional tool to be adopted for efficient and harmless SAP scaffold customization in tissue engineering.
Collapse
|
45
|
Fontana G, Delgado LM, Cigognini D. Biologically Inspired Materials in Tissue Engineering. EXTRACELLULAR MATRIX FOR TISSUE ENGINEERING AND BIOMATERIALS 2018. [DOI: 10.1007/978-3-319-77023-9_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
46
|
Zhuang P, Sun AX, An J, Chua CK, Chew SY. 3D neural tissue models: From spheroids to bioprinting. Biomaterials 2017; 154:113-133. [PMID: 29120815 DOI: 10.1016/j.biomaterials.2017.10.002] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/14/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022]
Abstract
Three-dimensional (3D) in vitro neural tissue models provide a better recapitulation of in vivo cell-cell and cell-extracellular matrix interactions than conventional two-dimensional (2D) cultures. Therefore, the former is believed to have great potential for both mechanistic and translational studies. In this paper, we review the recent developments in 3D in vitro neural tissue models, with a particular focus on the emerging bioprinted tissue structures. We draw on specific examples to describe the merits and limitations of each model, in terms of different applications. Bioprinting offers a revolutionary approach for constructing repeatable and controllable 3D in vitro neural tissues with diverse cell types, complex microscale features and tissue level responses. Further advances in bioprinting research would likely consolidate existing models and generate complex neural tissue structures bearing higher fidelity, which is ultimately useful for probing disease-specific mechanisms, facilitating development of novel therapeutics and promoting neural regeneration.
Collapse
Affiliation(s)
- Pei Zhuang
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.
| | - Alfred Xuyang Sun
- Department of Neurology, National Neuroscience Institute, 20 College Road, Singapore 169856, Singapore; Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore.
| | - Jia An
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.
| | - Chee Kai Chua
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|
47
|
Russell LN, Lampe KJ. Oligodendrocyte Precursor Cell Viability, Proliferation, and Morphology is Dependent on Mesh Size and Storage Modulus in 3D Poly(ethylene glycol)-Based Hydrogels. ACS Biomater Sci Eng 2017; 3:3459-3468. [PMID: 33445383 DOI: 10.1021/acsbiomaterials.7b00374] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oligodendrocytes in the central nervous system (CNS) are responsible for generating myelin, an electrically insulating layer around neuronal axons. When myelin is damaged, neurons are incapable of sustaining normal communications, which can manifest in patients as pain and loss of mobility and vision. A plethora of research has used biomaterials to promote neuronal regeneration, but despite the wide implications of a disrupted myelin sheath, very little is known about how biomaterial environments impact proliferation of oligodendrocyte precursor cells (OPCs) or their differentiation into myelinating oligodendrocytes. This work investigates how the storage modulus and mesh size of a polyethylene glycol (PEG)-based hydrogel, varied via two different mechanisms, directly affect the proliferation of two OPC lines encapsulated and cultured in 3D. Viability and proliferation of both OPC lines was dependent on hydrogel swelling and stiffness, where the concentration of ATP increased more in the more compliant gels. OPCs multiplied in the 3D hydrogels, creating significantly larger spheroids in the less cross-linked conditions. Stiffer, more highly cross-linked materials lead to greater expression of PDGFRα, an OPC receptor, indicating that fewer cells were committed to the oligodendrocyte lineage or had dedifferentiated in compliant materials. Laminin incorporation in the 3D matrix was found to have little effect on viability or proliferation. These findings provide valuable information on how mesh size and stiffness affect OPCs where more compliant materials favor proliferation of OPCs with less commitment to a mature oligodendrocyte lineage. Such information will be useful in the development of translational biomaterials to stimulate oligodendrocyte maturation for neural regeneration.
Collapse
Affiliation(s)
- Lauren N Russell
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, Virginia 22904, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, Virginia 22904, United States
| |
Collapse
|
48
|
Ayala-Caminero R, Pinzón-Herrera L, Martinez CAR, Almodovar J. Polymeric scaffolds for three-dimensional culture of nerve cells: a model of peripheral nerve regeneration. MRS COMMUNICATIONS 2017; 7:391-415. [PMID: 29515936 PMCID: PMC5836791 DOI: 10.1557/mrc.2017.90] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/28/2017] [Indexed: 05/09/2023]
Abstract
Understanding peripheral nerve repair requires the evaluation of 3D structures that serve as platforms for 3D cell culture. Multiple platforms for 3D cell culture have been developed, mimicking peripheral nerve growth and function, in order to study tissue repair or diseases. To recreate an appropriate 3D environment for peripheral nerve cells, key factors are to be considered including: selection of cells, polymeric biomaterials to be used, and fabrication techniques to shape and form the 3D scaffolds for cellular culture. This review focuses on polymeric 3D platforms used for the development of 3D peripheral nerve cell cultures.
Collapse
Affiliation(s)
- Radamés Ayala-Caminero
- Bioengineering Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayagüez, Puerto Rico, 00681-9000, USA
| | - Luis Pinzón-Herrera
- Department of Chemical Engineering, University of Puerto Rico Mayagüez, Call Box 9000, Mayaguez, Puerto Rico, 00681-9000, USA
| | - Carol A Rivera Martinez
- Bioengineering Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayagüez, Puerto Rico, 00681-9000, USA
| | - Jorge Almodovar
- Bioengineering Program, University of Puerto Rico Mayaguez, Call Box 9000, Mayagüez, Puerto Rico, 00681-9000, USA
| |
Collapse
|
49
|
Chen S, Zhou A, He B, Zhao W, Chen X, Jiang D. Designer D-form self-assembling peptide scaffolds promote the proliferation and migration of rat bone marrow-derived mesenchymal stem cells. Int J Mol Med 2017; 40:679-688. [PMID: 28677805 PMCID: PMC5547947 DOI: 10.3892/ijmm.2017.3056] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/23/2017] [Indexed: 01/06/2023] Open
Abstract
Self-assembling peptide (SAP) nanofiber hydrogel scaffolds have become increasingly important in tissue engineering due to their outstanding bioactivity and biodegradability. However, there is an initial concern on their long-term clinical use, since SAPs made of L-form amino acid sequences are sensitive to enzymatic degradation. In this study, we present a designer SAP, D-RADA16, made of all D-amino acid. We investigated the nanofiber morphology of D-RADA16, its potential for the culture of bone marrow-derived mesenchymal stem cells (BMSCs), and the proteolytic resistance of the biomaterial. The results revealed that D-RADA16 exhibited stable β-sheets and formed interwoven nanofiber scaffolds in water. D-RADA16 and L-RADA16 hydrogel scaffolds were both found to promote the proliferation and migration of rat BMSCs in the 3D cell culture microenvironment. Furthermore, the D-RADA16 scaffolds exhibited a higher proteolytic resistance against proteinase K than the L-RADA16 scaffolds. These observations indicate that D-RADA16 hydrogel scaffolds have excellent bioactivity, biocompatibility and biostability, and thus may serve as promising candidates for long-term application in vivo.
Collapse
Affiliation(s)
- Shuo Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Bin He
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaojun Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
50
|
Murphy AR, Laslett A, O'Brien CM, Cameron NR. Scaffolds for 3D in vitro culture of neural lineage cells. Acta Biomater 2017; 54:1-20. [PMID: 28259835 DOI: 10.1016/j.actbio.2017.02.046] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
Abstract
Understanding how neurodegenerative disorders develop is not only a key challenge for researchers but also for the wider society, given the rapidly aging populations in developed countries. Advances in this field require new tools with which to recreate neural tissue in vitro and produce realistic disease models. This in turn requires robust and reliable systems for performing 3D in vitro culture of neural lineage cells. This review provides a state of the art update on three-dimensional culture systems for in vitro development of neural tissue, employing a wide range of scaffold types including hydrogels, solid porous polymers, fibrous materials and decellularised tissues as well as microfluidic devices and lab-on-a-chip systems. To provide some context with in vivo development of the central nervous system (CNS), we also provide a brief overview of the neural stem cell niche, neural development and neural differentiation in vitro. We conclude with a discussion of future directions for this exciting and important field of biomaterials research. STATEMENT OF SIGNIFICANCE Neurodegenerative diseases, including dementia, Parkinson's and Alzheimer's diseases and motor neuron diseases, are a major societal challenge for aging populations. Understanding these conditions and developing therapies against them will require the development of new physical models of healthy and diseased neural tissue. Cellular models resembling neural tissue can be cultured in the laboratory with the help of 3D scaffolds - materials that allow the organization of neural cells into tissue-like structures. This review presents recent work on the development of different types of scaffolds for the 3D culture of neural lineage cells and the generation of functioning neural-like tissue. These in vitro culture systems are enabling the development of new approaches for modelling and tackling diseases of the brain and CNS.
Collapse
Affiliation(s)
- Ashley R Murphy
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC 3800, Australia
| | - Andrew Laslett
- CSIRO Manufacturing, Bag 10, Clayton South MDC, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | - Carmel M O'Brien
- CSIRO Manufacturing, Bag 10, Clayton South MDC, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | - Neil R Cameron
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC 3800, Australia.
| |
Collapse
|