1
|
Cui D, Guo W, Chang J, Fan S, Bai X, Li L, Yang C, Wang C, Li M, Fei J. Polydopamine-coated polycaprolactone/carbon nanotube fibrous scaffolds loaded with basic fibroblast growth factor for wound healing. Mater Today Bio 2024; 28:101190. [PMID: 39221197 PMCID: PMC11364907 DOI: 10.1016/j.mtbio.2024.101190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- Dapeng Cui
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Wei Guo
- Emergency Department, Peking University People's Hospital, Beijing, 100044, China
| | - Jing Chang
- Trauma Medicine Center, National Center for Trauma Medicine, Key Laboratory of Trauma and Neural Regeneration (Peking University, Ministry of Education), Peking University People's Hospital, Beijing, 100044, China
| | - Shuang Fan
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Xiaochen Bai
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Lei Li
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Chen Yang
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Chuanlin Wang
- Trauma Medicine Center, National Center for Trauma Medicine, Key Laboratory of Trauma and Neural Regeneration (Peking University, Ministry of Education), Peking University People's Hospital, Beijing, 100044, China
| | - Ming Li
- Trauma Medicine Center, National Center for Trauma Medicine, Key Laboratory of Trauma and Neural Regeneration (Peking University, Ministry of Education), Peking University People's Hospital, Beijing, 100044, China
| | - Jiandong Fei
- Hepatobiliary Surgery Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, 075000, China
| |
Collapse
|
2
|
Donati L, Valicenti ML, Giannoni S, Morena F, Martino S. Biomaterials Mimicking Mechanobiology: A Specific Design for a Specific Biological Application. Int J Mol Sci 2024; 25:10386. [PMID: 39408716 PMCID: PMC11476540 DOI: 10.3390/ijms251910386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Mechanosensing and mechanotransduction pathways between the Extracellular Matrix (ECM) and cells form the essential crosstalk that regulates cell homeostasis, tissue development, morphology, maintenance, and function. Understanding these mechanisms involves creating an appropriate cell support that elicits signals to guide cellular functions. In this context, polymers can serve as ideal molecules for producing biomaterials designed to mimic the characteristics of the ECM, thereby triggering responsive mechanisms that closely resemble those induced by a natural physiological system. The generated specific stimuli depend on the different natural or synthetic origins of the polymers, the chemical composition, the assembly structure, and the physical and surface properties of biomaterials. This review discusses the most widely used polymers and their customization to develop biomaterials with tailored properties. It examines how the characteristics of biomaterials-based polymers can be harnessed to replicate the functions of biological cells, making them suitable for biomedical and biotechnological applications.
Collapse
Affiliation(s)
- Leonardo Donati
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Maria Luisa Valicenti
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Samuele Giannoni
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
- Centro di Eccellenza Materiali Innovativi Nanostrutturati per Applicazioni Chimiche Fisiche e Biomediche (CEMIN), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
3
|
Chen Y, Li Y, Zhu W, Liu Q. Biomimetic gradient scaffolds for the tissue engineering and regeneration of rotator cuff enthesis. Biofabrication 2024; 16:032005. [PMID: 38697099 DOI: 10.1088/1758-5090/ad467d] [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: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Rotator cuff tear is one of the most common musculoskeletal disorders, which often results in recurrent shoulder pain and limited movement. Enthesis is a structurally complex and functionally critical interface connecting tendon and bone that plays an essential role in maintaining integrity of the shoulder joint. Despite the availability of advanced surgical procedures for rotator cuff repair, there is a high rate of failure following surgery due to suboptimal enthesis healing and regeneration. Novel strategies based on tissue engineering are gaining popularity in improving tendon-bone interface (TBI) regeneration. Through incorporating physical and biochemical cues into scaffold design which mimics the structure and composition of native enthesis is advantageous to guide specific differentiation of seeding cells and facilitate the formation of functional tissues. In this review, we summarize the current state of research in enthesis tissue engineering highlighting the development and application of biomimetic scaffolds that replicate the gradient TBI. We also discuss the latest techniques for fabricating potential translatable scaffolds such as 3D bioprinting and microfluidic device. While preclinical studies have demonstrated encouraging results of biomimetic gradient scaffolds, the translation of these findings into clinical applications necessitates a comprehensive understanding of their safety and long-term efficacy.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yexin Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Qian Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
4
|
Lemarié L, Dargar T, Grosjean I, Gache V, Courtial EJ, Sohier J. Human Induced Pluripotent Spheroids' Growth Is Driven by Viscoelastic Properties and Macrostructure of 3D Hydrogel Environment. Bioengineering (Basel) 2023; 10:1418. [PMID: 38136009 PMCID: PMC10740696 DOI: 10.3390/bioengineering10121418] [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: 11/13/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Stem cells, particularly human iPSCs, constitute a powerful tool for tissue engineering, notably through spheroid and organoid models. While the sensitivity of stem cells to the viscoelastic properties of their direct microenvironment is well-described, stem cell differentiation still relies on biochemical factors. Our aim is to investigate the role of the viscoelastic properties of hiPSC spheroids' direct environment on their fate. To ensure that cell growth is driven only by mechanical interaction, bioprintable alginate-gelatin hydrogels with significantly different viscoelastic properties were utilized in differentiation factor-free culture medium. Alginate-gelatin hydrogels of varying concentrations were developed to provide 3D environments of significantly different mechanical properties, ranging from 1 to 100 kPa, while allowing printability. hiPSC spheroids from two different cell lines were prepared by aggregation (⌀ = 100 µm, n > 1 × 104), included and cultured in the different hydrogels for 14 days. While spheroids within dense hydrogels exhibited limited growth, irrespective of formulation, porous hydrogels prepared with a liquid-liquid emulsion method displayed significant variations of spheroid morphology and growth as a function of hydrogel mechanical properties. Transversal culture (adjacent spheroids-laden alginate-gelatin hydrogels) clearly confirmed the separate effect of each hydrogel environment on hiPSC spheroid behavior. This study is the first to demonstrate that a mechanically modulated microenvironment induces diverse hiPSC spheroid behavior without the influence of other factors. It allows one to envision the combination of multiple formulations to create a complex object, where the fate of hiPSCs will be independently controlled by their direct microenvironment.
Collapse
Affiliation(s)
- Lucas Lemarié
- SEGULA Technologies, 69100 Villeurbanne, France;
- 3d.FAB, CNRS UMR 5246, ICBMS (Institute of Molecular and Supramolecular Chemistry and Biochemistry), Université Lyon 1, 69622 Villeurbanne, France;
- CNRS UMR 5305, LBTI (Tissue Biology and Therapeutic Engineering Laboratory), 69007 Lyon, France
| | - Tanushri Dargar
- CNRS UMR5261, INSERM U1315, INMG-PNMG (NeuroMyoGene Institute, Physiopathology and Genetics of the Neuron and the Muscle), Université Lyon 1, 69008 Lyon, France; (T.D.); (I.G.); (V.G.)
| | - Isabelle Grosjean
- CNRS UMR5261, INSERM U1315, INMG-PNMG (NeuroMyoGene Institute, Physiopathology and Genetics of the Neuron and the Muscle), Université Lyon 1, 69008 Lyon, France; (T.D.); (I.G.); (V.G.)
| | - Vincent Gache
- CNRS UMR5261, INSERM U1315, INMG-PNMG (NeuroMyoGene Institute, Physiopathology and Genetics of the Neuron and the Muscle), Université Lyon 1, 69008 Lyon, France; (T.D.); (I.G.); (V.G.)
| | - Edwin J. Courtial
- 3d.FAB, CNRS UMR 5246, ICBMS (Institute of Molecular and Supramolecular Chemistry and Biochemistry), Université Lyon 1, 69622 Villeurbanne, France;
| | - Jérôme Sohier
- CNRS UMR 5305, LBTI (Tissue Biology and Therapeutic Engineering Laboratory), 69007 Lyon, France
| |
Collapse
|
5
|
Murota Y, Nagane M, Wu M, Santra M, Venkateswaran S, Tanaka S, Bradley M, Taga T, Tabu K. A niche-mimicking polymer hydrogel-based approach to identify molecular targets for tackling human pancreatic cancer stem cells. Inflamm Regen 2023; 43:46. [PMID: 37759310 PMCID: PMC10523636 DOI: 10.1186/s41232-023-00296-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is one of the most fatal human cancers, but effective therapies remain to be established. Cancer stem cells (CSCs) are highly resistant to anti-cancer drugs and a deeper understanding of their microenvironmental niche has been considered important to provide understanding and solutions to cancer eradication. However, as the CSC niche is composed of a wide variety of biological and physicochemical factors, the development of multidisciplinary tools that recapitulate their complex features is indispensable. Synthetic polymers have been studied as attractive biomaterials due to their tunable biofunctionalities, while hydrogelation technique further renders upon them a diversity of physical properties, making them an attractive tool for analysis of the CSC niche. METHODS To develop innovative materials that recapitulate the CSC niche in pancreatic cancers, we performed polymer microarray analysis to identify niche-mimicking scaffolds that preferentially supported the growth of CSCs. The niche-mimicking activity of the identified polymers was further optimized by polyethylene glycol (PEG)-based hydrogelation. To reveal the biological mechanisms behind the activity of the optimized hydrogels towards CSCs, proteins binding onto the hydrogel were analyzed by liquid chromatography with tandem mass spectrometry (LC-MS/MS), and the potential therapeutic targets were validated by looking at gene expression and patients' outcome in the TCGA database. RESULTS PA531, a heteropolymer composed of 2-methoxyethyl methacrylate (MEMA) and 2-(diethylamino)ethyl methacrylate (DEAEMA) (5.5:4.5) that specifically supports the growth and maintenance of CSCs was identified by polymer microarray screening using the human PAAD cell line KLM1. The polymer PA531 was converted into five hydrogels (PA531-HG1 to HG5) and developed to give an optimized scaffold with the highest CSC niche-mimicking activities. From this polymer that recapitulated CSC binding and control, the proteins fetuin-B and angiotensinogen were identified as candidate target molecules with clinical significance due to the correlation between gene expression levels and prognosis in PAAD patients and the proteins associated with the niche-mimicking polymer. CONCLUSION This study screened for biofunctional polymers suitable for recapitulation of the pancreatic CSC niche and one hydrogel with high niche-mimicking abilities was successfully fabricated. Two soluble factors with clinical significance were identified as potential candidates for biomarkers and therapeutic targets in pancreatic cancers. Such a biomaterial-based approach could be a new platform in drug discovery and therapy development against CSCs, via targeting of their niche.
Collapse
Affiliation(s)
- Yoshitaka Murota
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan
| | - Mariko Nagane
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan
| | - Mei Wu
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Mithun Santra
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Seshasailam Venkateswaran
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Mark Bradley
- School of Chemistry, University of Edinburgh, Joseph Black Building, West Mains Road, Edinburgh, EH9 3FJ, UK
| | - Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan
| | - Kouichi Tabu
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-Ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
6
|
Hu J, Liu S, Fan C. Applications of functionally-adapted hydrogels in tendon repair. Front Bioeng Biotechnol 2023; 11:1135090. [PMID: 36815891 PMCID: PMC9934866 DOI: 10.3389/fbioe.2023.1135090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Despite all the efforts made in tissue engineering for tendon repair, the management of tendon injuries still poses a challenge, as current treatments are unable to restore the function of tendons following injuries. Hydrogels, due to their exceptional biocompatibility and plasticity, have been extensively applied and regarded as promising candidate biomaterials in tissue regeneration. Varieties of approaches have designed functionally-adapted hydrogels and combined hydrogels with other factors (e.g., bioactive molecules or drugs) or materials for the enhancement of tendon repair. This review first summarized the current state of knowledge on the mechanisms underlying the process of tendon healing. Afterward, we discussed novel strategies in fabricating hydrogels to overcome the issues frequently encountered during the applications in tendon repair, including poor mechanical properties and undesirable degradation. In addition, we comprehensively summarized the rational design of hydrogels for promoting stem-cell-based tendon tissue engineering via altering biophysical and biochemical factors. Finally, the role of macrophages in tendon repair and how they respond to immunomodulatory hydrogels were highlighted.
Collapse
Affiliation(s)
- Jiacheng Hu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Shen Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| |
Collapse
|
7
|
Calore AR, Srinivas V, Groenendijk L, Serafim A, Stancu IC, Wilbers A, Leoné N, Sanchez AA, Auhl D, Mota C, Bernaerts K, Harings JAW, Moroni L. Manufacturing of scaffolds with interconnected internal open porosity and surface roughness. Acta Biomater 2023; 156:158-176. [PMID: 35868592 DOI: 10.1016/j.actbio.2022.07.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/18/2023]
Abstract
Manufacturing of three-dimensional scaffolds with multiple levels of porosity are an advantage in tissue regeneration approaches to influence cell behavior. Three-dimensional scaffolds with surface roughness and intra-filament open porosity were successfully fabricated by additive manufacturing combined with chemical foaming and porogen leaching without the need of toxic solvents. The decomposition of sodium citrate, a chemical blowing agent, generated pores within the scaffold filaments, which were interconnected and opened to the external environment by leaching of a water-soluble sacrificial phase, as confirmed by micro-CT and buoyancy measurements. The additional porosity did not result in lower elastic modulus, but in higher strain at maximum load, i.e. scaffold ductility. Human mesenchymal stromal cells cultured for 24 h adhered in greater numbers on these scaffolds when compared to plain additive-manufactured ones, irrespectively of the scaffold pre-treatment method. Additionally, they showed a more spread and random morphology, which is known to influence cell fate. Cells cultured for a longer period exhibited enhanced metabolic activity while secreting higher osteogenic markers after 7 days in culture. STATEMENT OF SIGNIFICANCE: Inspired by the function of hierarchical cellular structures in natural materials, this work elucidates the development of scaffolds with multiscale porosity by combining in-situ foaming and additive manufacturing, and successive porogen leaching. The resulting scaffolds displayed enhanced mechanical toughness and multiscale pore network interconnectivity, combined with early differentiation of adult mesenchymal stromal cells into the osteogenic lineage.
Collapse
Affiliation(s)
- Andrea Roberto Calore
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration department, Maastricht University, Maastricht, the Netherlands; Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, the Netherlands
| | - Varun Srinivas
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, the Netherlands
| | - Linda Groenendijk
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, the Netherlands
| | - Andrada Serafim
- Advanced Polymer Materials Group, University Politehnica of Bucharest, Romania
| | | | | | - Nils Leoné
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration department, Maastricht University, Maastricht, the Netherlands
| | - Ane Albillos Sanchez
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration department, Maastricht University, Maastricht, the Netherlands
| | - Dietmar Auhl
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, the Netherlands; Polymerwerkstoffe und -technologien, Technische Universität Berlin, the Netherlands
| | - Carlos Mota
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration department, Maastricht University, Maastricht, the Netherlands
| | - Katrien Bernaerts
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, the Netherlands
| | - Jules A W Harings
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, the Netherlands.
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration department, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
8
|
Research progress of stem cell therapy for endometrial injury. Mater Today Bio 2022; 16:100389. [PMID: 36033375 PMCID: PMC9403503 DOI: 10.1016/j.mtbio.2022.100389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
Endometrial damage is an important factor leading to infertility and traditional conventional treatments have limited efficacy. As an emerging technology in recent years, stem cell therapy has provided new hope for the treatment of this disease. By comparing the advantages of stem cells from different sources, it is believed that menstrual blood endometrial stem cells have a good application prospect as a new source of stem cells. However, the clinical utility of stem cells is still limited by issues such as colonization rates, long-term efficacy, tumor formation, and storage and transportation. This paper summarizes the mechanism by which stem cells repair endometrial damage and clarifies the material basis of their effects from four aspects: replacement of damaged sites, paracrine effects, interaction with growth factors, and other new targets. According to the pathological characteristics and treatment requirements of intrauterine adhesion (IUA), the research work to solve the above problems from the aspects of functional bioscaffold preparation and multi-functional platform construction is also summarized. From the perspective of scaffold materials and component functions, this review will provide a reference for comprehensively optimizing the clinical application of stem cells.
Collapse
|
9
|
Guan S, Wang Y, Xie F, Wang S, Xu W, Xu J, Sun C. Carboxymethyl Chitosan and Gelatin Hydrogel Scaffolds Incorporated with Conductive PEDOT Nanoparticles for Improved Neural Stem Cell Proliferation and Neuronal Differentiation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238326. [PMID: 36500418 PMCID: PMC9740948 DOI: 10.3390/molecules27238326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
Tissue engineering scaffolds provide biological and physiochemical cures to guide tissue recovery, and electrical signals through the electroactive materials possess tremendous potential to modulate the cell fate. In this study, a novel electroactive hydrogel scaffold was fabricated by assembling poly(3,4-ethylenedioxythiophene) (PEDOT) nanoparticles on a carboxymethyl chitosan/gelatin (CMCS/Gel) composite hydrogel surface via in situ chemical polymerization. The chemical structure, morphology, conductivity, porosity, swelling rate, in vitro biodegradation, and mechanical properties of the prepared hydrogel samples were characterized. The adhesion, proliferation, and differentiation of neural stem cells (NSCs) on conductive hydrogels were investigated. The CMCS/Gel-PEDOT hydrogels exhibited high porosity, excellent water absorption, improved thermal stability, and adequate biodegradability. Importantly, the mechanical properties of the prepared hydrogels were similar to those of brain tissue, with electrical conductivity up to (1.52 ± 0.15) × 10-3 S/cm. Compared to the CMCS/Gel hydrogel, the incorporation of PEDOT nanoparticles significantly improved the adhesion of NSCs, and supported long-term cell growth and proliferation in a three-dimensional (3D) microenvironment. In addition, under the differentiation condition, the conductive hydrogel also significantly enhanced neuronal differentiation with the up-regulation of β-tubulin III expression. These results suggest that CMCS/Gel-PEDOT hydrogels may be an attractive conductive substrate for further studies on neural tissue repair and regeneration.
Collapse
Affiliation(s)
- Shui Guan
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
- Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China
- Correspondence: (S.G.); (J.X.); (C.S.)
| | - Yangbin Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Feng Xie
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shuping Wang
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Weiping Xu
- School of Ocean Science and Technology & Panjin Institute of Industrial Technology, Dalian University of Technology, Panjin 124221, China
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
- Correspondence: (S.G.); (J.X.); (C.S.)
| | - Changkai Sun
- Research & Educational Center for the Control Engineering of Translational Precision Medicine (R-ECCE-TPM), School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian 116024, China
- Correspondence: (S.G.); (J.X.); (C.S.)
| |
Collapse
|
10
|
Hong IS. Enhancing Stem Cell-Based Therapeutic Potential by Combining Various Bioengineering Technologies. Front Cell Dev Biol 2022; 10:901661. [PMID: 35865629 PMCID: PMC9294278 DOI: 10.3389/fcell.2022.901661] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/17/2022] [Indexed: 12/05/2022] Open
Abstract
Stem cell-based therapeutics have gained tremendous attention in recent years due to their wide range of applications in various degenerative diseases, injuries, and other health-related conditions. Therapeutically effective bone marrow stem cells, cord blood- or adipose tissue-derived mesenchymal stem cells (MSCs), embryonic stem cells (ESCs), and more recently, induced pluripotent stem cells (iPSCs) have been widely reported in many preclinical and clinical studies with some promising results. However, these stem cell-only transplantation strategies are hindered by the harsh microenvironment, limited cell viability, and poor retention of transplanted cells at the sites of injury. In fact, a number of studies have reported that less than 5% of the transplanted cells are retained at the site of injury on the first day after transplantation, suggesting extremely low (<1%) viability of transplanted cells. In this context, 3D porous or fibrous national polymers (collagen, fibrin, hyaluronic acid, and chitosan)-based scaffold with appropriate mechanical features and biocompatibility can be used to overcome various limitations of stem cell-only transplantation by supporting their adhesion, survival, proliferation, and differentiation as well as providing elegant 3-dimensional (3D) tissue microenvironment. Therefore, stem cell-based tissue engineering using natural or synthetic biomimetics provides novel clinical and therapeutic opportunities for a number of degenerative diseases or tissue injury. Here, we summarized recent studies involving various types of stem cell-based tissue-engineering strategies for different degenerative diseases. We also reviewed recent studies for preclinical and clinical use of stem cell-based scaffolds and various optimization strategies.
Collapse
Affiliation(s)
- In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Seongnam, South Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Seongnam, South Korea
- *Correspondence: In-Sun Hong,
| |
Collapse
|
11
|
A Molecular View on Biomaterials and Dental Stem Cells Interactions: Literature Review. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biomaterials and stem cells are essential components in the field of regenerative medicine. Various biomaterials have been designed that have appropriate biochemical and biophysical characteristics to mimic the microenvironment of an extracellular matrix. Dental stem cells (DT-MSCs) represent a novel source for the development of autologous therapies due to their easy availability. Although research on biomaterials and DT-MSCs has progressed, there are still challenges in the characteristics of biomaterials and the molecular mechanisms involved in regulating the behavior of DT-MSCs. In this review, the characteristics of biomaterials are summarized, and their classification according to their source, bioactivity, and different biological effects on the expansion and differentiation of DT-MSCs is summarized. Finally, advances in research on the interaction of biomaterials and the molecular components involved (mechanosensors and mechanotransduction) in DT-MSCs during their proliferation and differentiation are analyzed. Understanding the molecular dynamics of DT-MSCs and biomaterials can contribute to research in regenerative medicine and the development of autologous stem cell therapies.
Collapse
|
12
|
Mo C, Luo R, Chen Y. Advances in the stimuli-responsive injectable hydrogel for controlled release of drugs. Macromol Rapid Commun 2022; 43:e2200007. [PMID: 35344233 DOI: 10.1002/marc.202200007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/21/2022] [Indexed: 11/11/2022]
Abstract
The stimuli-responsiveness of injectable hydrogel has been drastically developed for the controlled release of drugs and achieved encouraging curative effects in a variety of diseases including wounds, cardiovascular diseases and tumors. The gelation, swelling and degradation of such hydrogels respond to endogenous biochemical factors (such as pH, reactive oxygen species, glutathione, enzymes, glucose) and/or to exogenous physical stimulations (like light, magnetism, electricity and ultrasound), thereby accurately releasing loaded drugs in response to specifically pathological status and as desired for treatment plan and thus improving therapeutic efficacy effectively. In this paper, we give a detailed introduction of recent progresses in responsive injectable hydrogels and focus on the design strategy of various stimuli-sensitivities and their resultant alteration of gel dissociation and drug liberation behaviour. Their application in disease treatment is also discussed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Chunxiang Mo
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, 410001, China
| | - Rui Luo
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, 410001, China
| | - Yuping Chen
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang, 410001, China
| |
Collapse
|
13
|
Voga M, Majdic G. Articular Cartilage Regeneration in Veterinary Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1401:23-55. [DOI: 10.1007/5584_2022_717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
14
|
Lee KY, Loh HX, Wan ACA. Systems for Muscle Cell Differentiation: From Bioengineering to Future Food. MICROMACHINES 2021; 13:71. [PMID: 35056236 PMCID: PMC8777594 DOI: 10.3390/mi13010071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022]
Abstract
In light of pressing issues, such as sustainability and climate change, future protein sources will increasingly turn from livestock to cell-based production and manufacturing activities. In the case of cell-based or cultured meat a relevant aspect would be the differentiation of muscle cells into mature muscle tissue, as well as how the microsystems that have been developed to date can be developed for larger-scale cultures. To delve into this aspect we review previous research that has been carried out on skeletal muscle tissue engineering and how various biological and physicochemical factors, mechanical and electrical stimuli, affect muscle cell differentiation on an experimental scale. Material aspects such as the different biomaterials used and 3D vs. 2D configurations in the context of muscle cell differentiation will also be discussed. Finally, the ability to translate these systems to more scalable bioreactor configurations and eventually bring them to a commercial scale will be touched upon.
Collapse
Affiliation(s)
| | | | - Andrew C. A. Wan
- Singapore Institute of Food and Biotechnology Innovation, 31 Biopolis Way, #01-02, Nanos, Singapore 138669, Singapore; (K.-Y.L.); (H.-X.L.)
| |
Collapse
|
15
|
Ozpinar EW, Frey AL, Cruse G, Freytes DO. Mast Cell-Biomaterial Interactions and Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:590-603. [PMID: 33164714 PMCID: PMC8739845 DOI: 10.1089/ten.teb.2020.0275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Tissue engineers often use biomaterials to provide structural support along with mechanical and chemical signals to modulate the wound healing process. Biomaterials that are implanted into the body interact with a heterogeneous and dynamic inflammatory environment that is present at the site of injury. Whether synthetically derived, naturally derived, or a combination of both, it is important to assess biomaterials for their ability to modulate inflammation to understand their potential clinical use. One important, but underexplored cell in the context of biomaterials is the mast cell (MC). MCs are granulocytic leukocytes that engage in a variety of events in both the innate and adaptive immune systems. Although highly recognized for their roles in allergic reactions, MCs play an important role in wound healing by recognizing antigens through pattern recognition receptors and the high-affinity immunoglobulin E receptor (FceRI) and releasing granules that affect cell recruitment, fibrosis, extracellular matrix deposition, angiogenesis, and vasculogenesis. MCs also mediate the foreign body response, contributing to the incorporation or rejection of implants. Studies of MC-biomaterial interactions can aid in the elucidation of MC roles during the host tissue response and tissue repair. This review is designed for those in the tissue engineering and biomaterial fields who are interested in exploring the role MCs may play in wound-biomaterial interactions and wound healing. With this review, we hope to inspire more research in the MC-biomaterial space to accelerate the design and construction of optimized implants. Impact statement Mast cells (MCs) are highly specialized inflammatory cells that have crucial, but not fully understood, roles in wound healing and tissue repair. Upon stimulation, they recognize foreign antigens and release granules that help orchestrate the inflammatory response after tissue damage or biomaterial implantation. This review summarizes the current use of MCs in biomaterial research along with literature from the past decade focusing on MC interactions with materials used for tissue repair and regeneration. Studying MC-biomaterial interactions will help (i) further understand the process of inflammation and (ii) design biomaterials and tissue-engineered constructs for optimal repair and regeneration.
Collapse
Affiliation(s)
- Emily W Ozpinar
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Ariana L Frey
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
| | - Glenn Cruse
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Donald O Freytes
- The Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina-Chapel Hill, Raleigh, North Carolina, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
16
|
Asl MA, Karbasi S, Beigi-Boroujeni S, Zamanlui Benisi S, Saeed M. Evaluation of the effects of starch on polyhydroxybutyrate electrospun scaffolds for bone tissue engineering applications. Int J Biol Macromol 2021; 191:500-513. [PMID: 34555400 DOI: 10.1016/j.ijbiomac.2021.09.078] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/02/2021] [Accepted: 09/12/2021] [Indexed: 11/25/2022]
Abstract
Efficient design for bone tissue engineering requires an understanding of the appropriate selection of biomimetic natural or synthetic materials and scalable fabrication technologies. In this research, poly (3-hydroxybutyrate) (PHB) and starch (5-15 wt%) as biological macromolecules were used to fabricate novel biomimetic scaffolds by electrospinning method. SEM results of electrospun scaffolds revealed bead-free nanofibers and three-dimensional homogenous structures with highly interconnected pores. Results of FTIR and Raman demonstrated that there were hydrogen bonds between the two polymers. The tensile strength of scaffolds was significantly improved by adding starch up to 10 wt%, from 3.05 to 15.54 MPa. In vitro degradation and hydrophilicity of the scaffolds were improved with the presence of starch. The viability and proliferation of MG-63 cells and alkaline phosphatase (ALP) activity were remarkably increased in the PHB-starch scaffolds compared to the PHB and control samples. The mineralization and calcium deposition of MG-63 cells were confirmed by alizarin red staining. It is concluded that PHB/starch electrospun scaffold could be a good candidate for bone tissue engineering applications.
Collapse
Affiliation(s)
- Maryam Abdollahi Asl
- Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran 1469669191, Iran
| | - Saeed Karbasi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Dental Implants Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Saeed Beigi-Boroujeni
- School of Engineering and Sciences, Tecnologico de Monterrey, Av. Eugenio Garza Sada Sur, Monterrey 2501, N. L., Mexico; Hard Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahdi Saeed
- Soft Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
17
|
Zhao X, Li Q, Guo Z, Li Z. Constructing a cell microenvironment with biomaterial scaffolds for stem cell therapy. Stem Cell Res Ther 2021; 12:583. [PMID: 34809719 PMCID: PMC8607654 DOI: 10.1186/s13287-021-02650-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/03/2021] [Indexed: 01/08/2023] Open
Abstract
Stem cell therapy is widely recognized as a promising strategy for exerting therapeutic effects after injury in degenerative diseases. However, limitations such as low cell retention and survival rates after transplantation exist in clinical applications. In recent years, emerging biomaterials that provide a supportable cellular microenvironment for transplanted cells have optimized the therapeutic efficacy of stem cells in injured tissues or organs. Advances in the engineered microenvironment are revolutionizing our understanding of stem cell-based therapies by co-transplanting with synthetic and tissue-derived biomaterials, which offer a scaffold for stem cells and propose an unprecedented opportunity to further employ significant influences in tissue repair and regeneration.
Collapse
Affiliation(s)
- Xiaotong Zhao
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China.,Department of Cardiology, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Qiong Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China. .,Department of Cardiology, Zhengzhou Seventh People's Hospital, Zhengzhou, China.
| | - Zongjin Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, Henan, China. .,Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
18
|
Shen CC, Yang MY, Chang KB, Tseng CH, Yang YP, Yang YC, Kung ML, Lai WY, Lin TW, Hsieh HH, Hung HS. Fabrication of hyaluronic acid-gold nanoparticles with chitosan to modulate neural differentiation of mesenchymal stem cells. J Chin Med Assoc 2021; 84:1007-1018. [PMID: 34320517 DOI: 10.1097/jcma.0000000000000589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Chitosan (Chi) is a natural material which has been widely used in neural applications due to possessing better biocompatibility. In this research study, a novel of nanocomposites film based on Chi with hyaluronic acid (HA), combined with varying amounts of gold nanoparticles (AuNPs), was created resulting in pure Chi, Chi-HA, Chi-HA-AuNPs (25 ppm), and Chi-HA-AuNPs (50 ppm). METHODS This study focused on evaluating their effects on mesenchymal stem cell (MSC) viability, colony formation, and biocompatibility. The surface morphology and chemical position were characterized through UV-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), SEM, and contact-angle assessment. RESULTS When seeding MSCs on Chi-HA-AuNPs (50 ppm), the results showed high cell viability, biocompatibility, and the highest colony formation ability. Meanwhile, the evidence showed that Chi-HA-Au nanofilm was able to inhibit nestin and β-tubulin expression of MSCs, as well as inhibit the ability of neurogenic differentiation. Furthermore, the results of matrix metalloproteinase 2/9 (MMP2/9) expression in MSCs were also significantly higher in the Chi-HA-AuNP (50 ppm) group, guiding with angiogenesis and wound healing abilities. In addition, in our rat model, both capsule thickness and collagen deposition were the lowest in Chi-HA-AuNPs (50 ppm). CONCLUSION Thus, in view of the in vitro and in vivo results, Chi-HA-AuNPs (50 ppm) could not only maintain the greatest stemness properties and regulate the neurogenic differentiation ability of MSCs, but was able to also induce the least immune response. Herein, Chi-HA-Au 50 ppm nanofilm holds promise as a suitable material for nerve regeneration engineering.
Collapse
Affiliation(s)
- Chiung-Chyi Shen
- Neurological Institute Head of Department of Neurosurgery Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Department of Physical Therapy, Hung Kuang University, Taichung, Taiwan, ROC
- Basic Medical Education Center, Central Taiwan University of Science and Technology, Taichung, Taiwan, ROC
| | - Meng-Yin Yang
- Neurological Institute Head of Department of Neurosurgery Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Basic Medical Education Center, Central Taiwan University of Science and Technology, Taichung, Taiwan, ROC
| | - Kai-Bo Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan, ROC
| | - Chia-Hsuan Tseng
- Department of Occupational Safety and Health, China Medical University, Taichung, Taiwan, ROC
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Chin Yang
- Neurological Institute Head of Department of Neurosurgery Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | - Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Tzu-Wei Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsien-Hsu Hsieh
- Blood Bank, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | - Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan, ROC
- Translational Medicine Research, China Medical University Hospital, Taichung, Taiwan, ROC
| |
Collapse
|
19
|
Hung HS, Kao WC, Shen CC, Chang KB, Tang CM, Yang MY, Yang YC, Yeh CA, Li JJ, Hsieh HH. Inflammatory Modulation of Polyethylene Glycol-AuNP for Regulation of the Neural Differentiation Capacity of Mesenchymal Stem Cells. Cells 2021; 10:2854. [PMID: 34831077 PMCID: PMC8616252 DOI: 10.3390/cells10112854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
A nanocomposite composed of polyethylene glycol (PEG) incorporated with various concentrations (~17.4, ~43.5, ~174 ppm) of gold nanoparticles (Au) was created to investigate its biocompatibility and biological performance in vitro and in vivo. First, surface topography and chemical composition was determined through UV-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), atomic force microscopy (AFM), scanning electron microscopy (SEM), free radical scavenging ability, and water contact angle measurement. Additionally, the diameters of the PEG-Au nanocomposites were also evaluated through dynamic light scattering (DLS) assay. According to the results, PEG containing 43.5 ppm of Au demonstrated superior biocompatibility and biological properties for mesenchymal stem cells (MSCs), as well as superior osteogenic differentiation, adipocyte differentiation, and, particularly, neuronal differentiation. Indeed, PEG-Au 43.5 ppm induced better cell adhesion, proliferation and migration in MSCs. The higher expression of the SDF-1α/CXCR4 axis may be associated with MMPs activation and may have also promoted the differentiation capacity of MSCs. Moreover, it also prevented MSCs from apoptosis and inhibited macrophage and platelet activation, as well as reactive oxygen species (ROS) generation. Furthermore, the anti-inflammatory, biocompatibility, and endothelialization capacity of PEG-Au was measured in a rat model. After implanting the nanocomposites into rats subcutaneously for 4 weeks, PEG-Au 43.5 ppm was able to enhance the anti-immune response through inhibiting CD86 expression (M1 polarization), while also reducing leukocyte infiltration (CD45). Moreover, PEG-Au 43.5 ppm facilitated CD31 expression and anti-fibrosis ability. Above all, the PEG-Au nanocomposite was evidenced to strengthen the differentiation of MSCs into various cells, including fat, vessel, and bone tissue and, particularly, nerve cells. This research has elucidated that PEG combined with the appropriate amount of Au nanoparticles could become a potential biomaterial able to cooperate with MSCs for tissue regeneration engineering.
Collapse
Affiliation(s)
- Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
- Translational Medicine Research, China Medical University Hospital, Taichung 40402, Taiwan
| | - Wei-Chien Kao
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Chiung-Chyi Shen
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (M.-Y.Y.); (Y.-C.Y.)
- Department of Physical Therapy, Hung Kuang University, Taichung 433304, Taiwan
- Basic Medical Education Center, Central Taiwan University of Science and Technology, Taichung 406053, Taiwan
| | - Kai-Bo Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Cheng-Ming Tang
- College of Oral Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Blood Bank, Taichung Veterans General Hospital, Taichung 407204, Taiwan;
| | - Meng-Yin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (M.-Y.Y.); (Y.-C.Y.)
| | - Yi-Chin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (M.-Y.Y.); (Y.-C.Y.)
| | - Chun-An Yeh
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Jia-Jhan Li
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Hsien-Hsu Hsieh
- Blood Bank, Taichung Veterans General Hospital, Taichung 407204, Taiwan;
| |
Collapse
|
20
|
Khazaei S, Soleimani M, Tafti SHA, Aghdam RM, Hojati Z. Improvement of Heart Function After Transplantation of Encapsulated Stem Cells Induced with miR-1/Myocd in Myocardial Infarction Model of Rat. Cell Transplant 2021; 30:9636897211048786. [PMID: 34606735 PMCID: PMC8493326 DOI: 10.1177/09636897211048786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular disease is one of the most common causes of death worldwide. Mesenchymal stem cells (MSCs) are one of the most common sources in cell-based therapies in heart regeneration. There are several methods to differentiate MSCs into cardiac-like cells, such as gene induction. Moreover, using a three-dimensional (3D) culture, such as hydrogels increases efficiency of differentiation. In the current study, mouse adipose-derived MSCs were co-transduced with lentiviruses containing microRNA-1 (miR-1) and Myocardin (Myocd). Then, expression of cardiac markers, such as NK2 homeobox 5(Nkx2-5), GATA binding protein 4 (Gata4), and troponin T type 2 (Tnnt2) was investigated, at both gene and protein levels in two-dimensional (2D) culture and chitosan/collagen hydrogel (CS/CO) as a 3D culture. Additionally, after induction of myocardial infarction (MI) in rats, a patch containing the encapsulated induced cardiomyocytes (iCM/P) was implanted to MI zone. Subsequently, 30 days after MI induction, echocardiography, immunohistochemistry staining, and histological examination were performed to evaluate cardiac function. The results of quantitative real -time polymerase chain reaction (qRT-PCR) and immunocytochemistry showed that co-induction of miR-1 and Myocd in MSCs followed by 3D culture of transduced cells increased expression of cardiac markers. Besides, results of in vivo study implicated that heart function was improved in MI model of rats in iCM/P-treated group. The results suggested that miR-1/Myocd induction combined with encapsulation of transduced cells in CS/CO hydrogel increased efficiency of MSCs differentiation into iCMs and could improve heart function in MI model of rats after implantation.
Collapse
Affiliation(s)
- Samaneh Khazaei
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, Isfahan University, Isfahan, Iran
| | - Masoud Soleimani
- Tissue Engineering and Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Tissue Engineering and Nanomedicine Research Center, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zohreh Hojati
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, Isfahan University, Isfahan, Iran
| |
Collapse
|
21
|
Interfacial Compatibilization into PLA/Mg Composites for Improved In Vitro Bioactivity and Stem Cell Adhesion. Molecules 2021; 26:molecules26195944. [PMID: 34641488 PMCID: PMC8512483 DOI: 10.3390/molecules26195944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 01/22/2023] Open
Abstract
The present work highlights the crucial role of the interfacial compatibilization on the design of polylactic acid (PLA)/Magnesium (Mg) composites for bone regeneration applications. In this regard, an amphiphilic poly(ethylene oxide-b-L,L-lactide) diblock copolymer with predefined composition was synthesised and used as a new interface to provide physical interactions between the metallic filler and the biopolymer matrix. This strategy allowed (i) overcoming the PLA/Mg interfacial adhesion weakness and (ii) modulating the composite hydrophilicity, bioactivity and biological behaviour. First, a full study of the influence of the copolymer incorporation on the morphological, wettability, thermal, thermo-mechanical and mechanical properties of PLA/Mg was investigated. Subsequently, the bioactivity was assessed during an in vitro degradation in simulated body fluid (SBF). Finally, biological studies with stem cells were carried out. The results showed an increase of the interfacial adhesion by the formation of a new interphase between the hydrophobic PLA matrix and the hydrophilic Mg filler. This interface stabilization was confirmed by a decrease in the damping factor (tanδ) following the copolymer addition. The latter also proves the beneficial effect of the composite hydrophilicity by selective surface localization of the hydrophilic PEO leading to a significant increase in the protein adsorption. Furthermore, hydroxyapatite was formed in bulk after 8 weeks of immersion in the SBF, suggesting that the bioactivity will be noticeably improved by the addition of the diblock copolymer. This ceramic could react as a natural bonding junction between the designed implant and the fractured bone during osteoregeneration. On the other hand, a slight decrease of the composite mechanical performances was noted.
Collapse
|
22
|
Lomboni DJ, Steeves A, Schock S, Bonetti L, De Nardo L, Variola F. Compounded topographical and physicochemical cueing by micro-engineered chitosan substrates on rat dorsal root ganglion neurons and human mesenchymal stem cells. SOFT MATTER 2021; 17:5284-5302. [PMID: 34075927 DOI: 10.1039/d0sm02170a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Given the intertwined physicochemical effects exerted in vivo by both natural and synthetic (e.g., biomaterial) interfaces on adhering cells, the evaluation of structure-function relationships governing cellular response to micro-engineered surfaces for applications in neuronal tissue engineering requires the use of in vitro testing platforms which consist of a clinically translatable material with tunable physiochemical properties. In this work, we micro-engineered chitosan substrates with arrays of parallel channels with variable width (20 and 60 μm). A citric acid (CA)-based crosslinking approach was used to provide an additional level of synergistic cueing on adhering cells by regulating the chitosan substrate's stiffness. Morphological and physicochemical characterization was conducted to unveil the structure-function relationships which govern the activity of rat dorsal root ganglion neurons (DRGs) and human mesenchymal stem cells (hMSCs), ultimately singling out the key role of microtopography, roughness and substrate's stiffness. While substrate's stiffness predominantly affected hMSC spreading, the modulation of the channels' design affected the neuronal architecture's complexity and guided the morphological transition of hMSCs. Finally, the combined analysis of tubulin expression and cell morphology allowed us to cast new light on the predominant role of the microtopography over substrate's stiffness in the process of hMSCs neurogenic differentiation.
Collapse
Affiliation(s)
- David J Lomboni
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Alexander Steeves
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Sarah Schock
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada and The Children's Hospital of Eastern Ontario (CHEO) Research Institute, Canada
| | - Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering, "G. Natta", Politecnico di Milano, Italy
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering, "G. Natta", Politecnico di Milano, Italy
| | - Fabio Variola
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada and Department of Cellular and Molecular Medicine, University of Ottawa, Canada and The Children's Hospital of Eastern Ontario (CHEO) Research Institute, Canada
| |
Collapse
|
23
|
Lin Y, Zhang L, Yang Y, Yang M, Hong Q, Chang K, Dai J, Chen L, Pan C, Hu Y, Quan L, Wei Y, Liu S, Yang Z. Loading Gentamicin and Zn 2+ on TiO 2 Nanotubes to Improve Anticoagulation, Endothelial Cell Growth, and Antibacterial Activities. Stem Cells Int 2021; 2021:9993247. [PMID: 34054972 PMCID: PMC8112940 DOI: 10.1155/2021/9993247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/05/2022] Open
Abstract
Titanium and its alloys are widely used in blood-contacting implantable and interventional medical devices; however, their biocompatibility is still facing great challenges. In the present study, in order to improve the biocompatibility and antibacterial activities of titanium, TiO2 nanotubes were firstly in situ prepared on the titanium surface by anodization, followed by the introduction of polyacrylic acid (PAA) and gentamicin (GS) on the nanotube surface by layer-by-layer assembly, and finally, zinc ions were loaded on the surface to further improve the bioactivities. The nanotubes displayed excellent hydrophilicity and special nanotube-like structure, which can selectively promote the albumin adsorption, enhance the blood compatibility, and promote the growth of endothelial cells to some degree. After the introduction of PAA and GS, although the superhydrophilicity cannot be achieved, the results of platelet adhesion, cyclic guanosine monophosphate (cGMP) activity, hemolysis rate, and activated partial thromboplastin time (APTT) showed that the blood compatibility was improved, and the blood compatibility was further enhanced after zinc ion loading. On the other hand, the modified surface showed good cytocompatibility to endothelial cells. The introduction of PAA and zinc ions not only promoted the adhesion and proliferation of endothelial cells but also upregulated expression of vascular endothelial growth factor (VEGF) and nitric oxide (NO). The slow and continuous release of GS and Zn2+ over 14 days can significantly improve the antibacterial properties. Therefore, the present study provides an effective method for the surface modification of titanium-based blood-contacting materials to simultaneously endow with good blood compatibility, endothelial growth behaviors, and antibacterial properties.
Collapse
Affiliation(s)
- Yuebin Lin
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Li Zhang
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Ya Yang
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Minhui Yang
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Qingxiang Hong
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Keming Chang
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Juan Dai
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Lu Chen
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Changjiang Pan
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Youdong Hu
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Li Quan
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Yanchun Wei
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Sen Liu
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Zhongmei Yang
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| |
Collapse
|
24
|
Esdaille CJ, Washington KS, Laurencin CT. Regenerative engineering: a review of recent advances and future directions. Regen Med 2021; 16:495-512. [PMID: 34030463 PMCID: PMC8356698 DOI: 10.2217/rme-2021-0016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Regenerative engineering is defined as the convergence of the disciplines of advanced material science, stem cell science, physics, developmental biology and clinical translation for the regeneration of complex tissues and organ systems. It is an expansion of tissue engineering, which was first developed as a method of repair and restoration of human tissue. In the past three decades, advances in regenerative engineering have made it possible to treat a variety of clinical challenges by utilizing cutting-edge technology currently available to harness the body's healing and regenerative abilities. The emergence of new information in developmental biology, stem cell science, advanced material science and nanotechnology have provided promising concepts and approaches to regenerate complex tissues and structures.
Collapse
Affiliation(s)
- Caldon J Esdaille
- Howard University College of Medicine, Washington, DC 20011, USA
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond & Beverly Sackler Center for Biomedical, Biological, Physical & Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
| | - Kenyatta S Washington
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond & Beverly Sackler Center for Biomedical, Biological, Physical & Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond & Beverly Sackler Center for Biomedical, Biological, Physical & Engineering Sciences, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06030, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, USA
| |
Collapse
|
25
|
Argentati C, Morena F, Fontana C, Tortorella I, Emiliani C, Latterini L, Zampini G, Martino S. Functionalized Silica Star-Shaped Nanoparticles and Human Mesenchymal Stem Cells: An In Vitro Model. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:779. [PMID: 33803869 PMCID: PMC8003255 DOI: 10.3390/nano11030779] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022]
Abstract
The biomedical translational applications of functionalized nanoparticles require comprehensive studies on their effect on human stem cells. Here, we have tested neat star-shaped mesoporous silica nanoparticles (s-MSN) and their chemically functionalized derivates; we examined nanoparticles (NPs) with similar dimensions but different surface chemistry, due to the amino groups grafted on silica nanoparticles (s-MSN-NH2), and gold nanoseeds chemically adsorbed on silica nanoparticles (s-MSN-Au). The different samples were dropped on glass coverslips to obtain a homogeneous deposition differing only for NPs' chemical functionalization and suitable for long-term culture of human Bone Marrow-Mesenchymal stem cells (hBM-MSCs) and Adipose stem cells (hASCs). Our model allowed us to demonstrate that hBM-MSCs and hASCs have comparable growth curves, viability, and canonical Vinculin Focal adhesion spots on functionalized s-MSN-NH2 and s-MSN-Au as on neat s-MSN and control systems, but also to show morphological changes on all NP types compared to the control counterparts. The new shape was stem-cell-specific and was maintained on all types of NPs. Compared to the other NPs, s-MSN-Au exerted a small genotoxic effect on both stem cell types, which, however, did not affect the stem cell behavior, likely due to a peculiar stem cell metabolic restoration response.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (C.A.); (F.M.); (I.T.); (C.E.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (C.A.); (F.M.); (I.T.); (C.E.)
| | - Chiara Fontana
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy; (C.F.); (L.L.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (C.A.); (F.M.); (I.T.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (C.A.); (F.M.); (I.T.); (C.E.)
| | - Loredana Latterini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy; (C.F.); (L.L.)
| | - Giulia Zampini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy; (C.F.); (L.L.)
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (C.A.); (F.M.); (I.T.); (C.E.)
| |
Collapse
|
26
|
Hashemzadeh MR, Taghavizadeh Yazdi ME, Amiri MS, Mousavi SH. Stem cell therapy in the heart: Biomaterials as a key route. Tissue Cell 2021; 71:101504. [PMID: 33607524 DOI: 10.1016/j.tice.2021.101504] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are one of the main concerns, nowadays causing a high rate of mortality in the world. The majority of conventional treatment protects the heart from failure progression. As a novel therapeutic way, Regenerative medicine in the heart includes cellular and noncellular approaches. Despite the irrefutable privileges of noncellular aspects such as administration of exosomes, utilizing of miRNAs, and growth factors, they cannot reverse necrotic or ischemic myocardium, hence recruiting of stem cells to help regenerative therapy in the heart seems indispensable. Stem cell lineages are varied and divided into two main groups namely pluripotent and adult stem cells. Not only has each of which own regenerative capacity, benefits, and drawbacks, but their turnover also close correlates with the target organ and/or tissue as well as the stage and level of failure. In addition to inefficient tissue integration due to the defects in delivering methods and poor retention of transplanted cells, the complexity of the heart and its movement also make more rigorous the repair process. Hence, utilizing biomaterials can make a key route to tackle such obstacles. In this review, we evaluate some natural products which can help stem cells in regenerative medicine of the cardiovascular system.
Collapse
Affiliation(s)
- Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad, Iran; Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Chen H, Zhang Y, Ni T, Ding P, Zan Y, Cai X, Zhang Y, Liu M, Pei R. Construction of a Silk Fibroin/Polyethylene Glycol Double Network Hydrogel with Co-Culture of HUVECs and UCMSCs for a Functional Vascular Network. ACS APPLIED BIO MATERIALS 2021; 4:406-419. [PMID: 35014292 DOI: 10.1021/acsabm.0c00353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The success of complex tissue and internal organ reconstruction relies principally on the fabrication of a 3D vascular network, which guarantees the delivery of oxygen and nutrients in addition to the disposal of waste. In this study, a rapidly forming cell-encapsulated double network (DN) hydrogel is constructed by an ultrasonically activated silk fibroin network and bioorthogonal-mediated polyethylene glycol network. This DN hydrogel can be solidified within 10 s, and its mechanical property gradually increases to ∼20 kPa after 30 min. This work also demonstrates that coencapsulation of human umbilical vein endothelial cells (HUVECs) and umbilical cord-derived mesenchymal stem cells (UCMSCs) into the DN hydrogel can facilitate the formation of more mature vessels and complete the capillary network in comparison with the hydrogels encapsulated with a single cell type both in vitro and in vivo. Taking together, the DN hydrogel, combined with coencapsulation of HUVECs and UCMSCs, represents a strategy for the construction of a functional vascular network.
Collapse
Affiliation(s)
- Hong Chen
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.,School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China.,Institut de Science des Matériaux de Mulhouse, IS2M-UMR CNRS 7361, UHA, 15, Rue Jean Starcky, Cedex 68057 Mulhouse, France
| | - Yajie Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tianyu Ni
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Pi Ding
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yue Zan
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.,School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xue Cai
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.,The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou 215004, China
| | - Yiwei Zhang
- Institute for Interdisciplinary Research, Jianghan University, Wuhan 430056, China
| | - Min Liu
- Institute for Interdisciplinary Research, Jianghan University, Wuhan 430056, China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
28
|
Padalhin A, Ventura R, Kim B, Sultana T, Park CM, Lee BT. Boosting osteogenic potential and bone regeneration by co-cultured cell derived extracellular matrix incorporated porous electrospun scaffold. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:779-798. [PMID: 33375905 DOI: 10.1080/09205063.2020.1869879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Implants for bone regeneration to remedy segmental bone defects, osteomyelitis, necrotic bone tissue and non-union fractures have worldwide appeal. Although biomaterials offer most of the advantages by improving tissue growth but developments are more commonly achieved via biologically derived molecules. To aid site specific bone tissue regeneration by synthetic scaffold, cell derived extracellular matrix (ECM) can be a crucial component. In this study, co-cultured bone marrow mesenchymal stem cell and osteoblastic cells derived ECM incorporated electrospun polycaprolactone (PCL) membranes were assessed for bone tissue engineering application. The preliminary experimental details indicated that, co-culture of cells supported enhanced in vitro ECM synthesis followed by successful deposition of osteoblastic ECM into electrospun membranes. The acellular samples revealed retention of ECM related biomacromolecules (collagen, glycosaminoglycan) and partial recovery of pores after decellularization. In vitro biocompatibility tests ensured improvement of proliferation and osteoblastic differentiation of MC3T3-E1 cells in decellularized ECM containing membrane (PCL-ECM) compared to bare membrane (PCL-B) which was further confirmed by osteogenic marker proteins expression analysis. The decellularized PCL-ECM membrane allowed great improvement of bone regeneration over the bare membrane (PCL-B) in 8 mm size critical sized rat skull defects at 2 months of post implantation. In short, the outcome of this study could be impactful in development and application of cell derived ECM based synthetic electrospun templates for bone tissue engineering application.[Formula: see text].
Collapse
Affiliation(s)
- Andrew Padalhin
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Reiza Ventura
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Boram Kim
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Tamanna Sultana
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| | - Chan Mi Park
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
29
|
Morena F, Argentati C, Soccio M, Bicchi I, Luzi F, Torre L, Munari A, Emiliani C, Gigli M, Lotti N, Armentano I, Martino S. Unpatterned Bioactive Poly(Butylene 1,4-Cyclohexanedicarboxylate)-Based Film Fast Induced Neuronal-Like Differentiation of Human Bone Marrow-Mesenchymal Stem Cells. Int J Mol Sci 2020; 21:E9274. [PMID: 33291757 PMCID: PMC7729499 DOI: 10.3390/ijms21239274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/19/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Herein, we present poly(butylene 1,4-cyclohexanedicarboxylate) (PBCE) films characterized by an unpatterned microstructure and a specific hydrophobicity, capable of boosting a drastic cytoskeleton architecture remodeling, culminating with the neuronal-like differentiation of human bone marrow-mesenchymal stem cells (hBM-MSCs). We have used two different filming procedures to prepare the films, solvent casting (PBCE) and compression-moulding (PBCE*). PBCE film had a rough and porous surface with spherulite-like aggregations (Ø = 10-20 μm) and was characterized by a water contact angle = 100°. PBCE* showed a smooth and continuous surface without voids and visible spherulite-like aggregations and was more hydrophobic (WCA = 110°). Both surface characteristics were modulated through the copolymerization of different amounts of ether-oxygen-containing co-units into PBCE chemical structure. We showed that only the surface characteristics of PBCE-solvent-casted films steered hBM-MSCs toward a neuronal-like differentiation. hBM-MSCs lost their canonical mesenchymal morphology, acquired a neuronal polarized shape with a long cell protrusion (≥150 μm), expressed neuron-specific class III β-tubulin and microtubule-associated protein 2 neuronal markers, while nestin, a marker of uncommitted stem cells, was drastically silenced. These events were observed as early as 2-days after cell seeding. Of note, the phenomenon was totally absent on PBCE* film, as hBM-MSCs maintained the mesenchymal shape and behavior and did not express neuronal/glial markers.
Collapse
Affiliation(s)
- Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (F.M.); (C.A.); (I.B.); (C.E.)
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (F.M.); (C.A.); (I.B.); (C.E.)
| | - Michelina Soccio
- Department of Civil, Chemical, Environmental, and Materials Engineering–DICAM, University of Bologna, 40136 Bologna, Italy; (M.S.); (A.M.)
| | - Ilaria Bicchi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (F.M.); (C.A.); (I.B.); (C.E.)
| | - Francesca Luzi
- Civil and Environmental Engineering Department, UdR INSTM, University of Perugia, 05100 Terni, Italy; (F.L.); (L.T.)
| | - Luigi Torre
- Civil and Environmental Engineering Department, UdR INSTM, University of Perugia, 05100 Terni, Italy; (F.L.); (L.T.)
| | - Andrea Munari
- Department of Civil, Chemical, Environmental, and Materials Engineering–DICAM, University of Bologna, 40136 Bologna, Italy; (M.S.); (A.M.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (F.M.); (C.A.); (I.B.); (C.E.)
- CEMIN, University of Perugia, 06123 Perugia, Italy
| | - Matteo Gigli
- Department of Molecular Sciences and Nanosystems, Ca’Foscari University of Venice, 30170 Venezia Mestre, Italy;
| | - Nadia Lotti
- Department of Civil, Chemical, Environmental, and Materials Engineering–DICAM, University of Bologna, 40136 Bologna, Italy; (M.S.); (A.M.)
| | - Ilaria Armentano
- Department of Economics, Engineering, Society and Business Organization (DEIM), University of Tuscia, 01100 Viterbo, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy; (F.M.); (C.A.); (I.B.); (C.E.)
- CEMIN, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
30
|
Luzi F, Tortorella I, Di Michele A, Dominici F, Argentati C, Morena F, Torre L, Puglia D, Martino S. Novel Nanocomposite PLA Films with Lignin/Zinc Oxide Hybrids: Design, Characterization, Interaction with Mesenchymal Stem Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2176. [PMID: 33142867 PMCID: PMC7692172 DOI: 10.3390/nano10112176] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 01/16/2023]
Abstract
Herein we present the production of novel nanocomposite films consisting of polylactic acid (PLA) polymer and the inclusion of nanoparticles of lignin (LNP), ZnO and hybrid ZnO@LNP (ZnO, 3.5% wt, ICP), characterized by similar regular shapes and different diameter distribution (30-70 nm and 100-150 nm, respectively). The obtained set of binary, ternary and quaternary systems were similar in surface wettability and morphology but different in the tensile performance: while the presence of LNP and ZnO in PLA caused a reduction of elastic modulus, stress and deformation at break, the inclusion of ZnO@LNP increased the stiffness and tensile strength (σb = 65.9 MPa and EYoung = 3030 MPa) with respect to neat PLA (σb = 37.4 MPa and EYoung = 2280 MPa). Neat and nanocomposite PLA-derived films were suitable for adult human bone marrow-mesenchymal stem cells and adipose stem cell cultures, as showed by their viability and behavior comparable to control conditions. Both stem cell types adhered to the films' surface by vinculin focal adhesion spots and responded to the films' mechanical properties by orchestrating the F-actin-filamin A interaction. Collectively, our results support the biomedical application of neat- and nanocomposite-PLA films and, based on the absence of toxicity in seeded stem cells, provide a proof of principle of their safety for food packaging purposes.
Collapse
Affiliation(s)
- Francesca Luzi
- Department of Civil and Environmental Engineering, Materials Engineering Center, UdR INSTM, University of Perugia, Strada di Pentima 4, 05100 Terni, Italy; (F.L.); (F.D.); (L.T.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (I.T.); (C.A.); (F.M.)
| | - Alessandro Di Michele
- Department of Physics and Geology, University of Perugia, Via Pascoli, 1, 06123 Perugia, Italy;
| | - Franco Dominici
- Department of Civil and Environmental Engineering, Materials Engineering Center, UdR INSTM, University of Perugia, Strada di Pentima 4, 05100 Terni, Italy; (F.L.); (F.D.); (L.T.)
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (I.T.); (C.A.); (F.M.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (I.T.); (C.A.); (F.M.)
| | - Luigi Torre
- Department of Civil and Environmental Engineering, Materials Engineering Center, UdR INSTM, University of Perugia, Strada di Pentima 4, 05100 Terni, Italy; (F.L.); (F.D.); (L.T.)
| | - Debora Puglia
- Department of Civil and Environmental Engineering, Materials Engineering Center, UdR INSTM, University of Perugia, Strada di Pentima 4, 05100 Terni, Italy; (F.L.); (F.D.); (L.T.)
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (I.T.); (C.A.); (F.M.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06123 Perugia, Italy
| |
Collapse
|
31
|
Mohamed Abudhahir K, Murugesan R, Vijayashree R, Selvamurugan N, Chung TW, Moorthi A. Metal doped calcium silicate biomaterial for skin tissue regeneration in vitro. J Biomater Appl 2020; 36:140-151. [PMID: 33050835 DOI: 10.1177/0885328220962607] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This study spots light on combined Wound healing process conjoining blood coagulation, inflammation reduction, proliferation and remodeling of the cells. The objective is to overcome the drawbacks of conventional clinically applied wound dressings such as poor rigidity, porosity, mechanical potency and bactericidal activity. As nosocomial infection is a very common condition at the wound site, bio-adhesive materials with intrinsic antibacterial properties are used in clinical applications. Considering the provenability of Wollastonite [Calcium silicate (CaSiO3)] to regenerate the soft tissues by inducing vascularization and regeneration of fibroblast cells And the antibacterial potentiality of zinc in clinical applications, the present study focuses on synthesis of Zn-Ws particles and evaluation of its antimicrobial and wound healing potentialities towards skin tissue engineering applications. The compositional characterization by EDAS and FT-IR spectral analysis have substantiated the presence of major elements and corresponding band stretching associated with the synthesized particles whereas the particles morphology by SEM images have shown the size of the Ws and Zn-Ws to be 370 nm and 530 nm respectively. From the in vitro studies, skin regenerative potential of Zn-Ws was determined on promoting fibroblast cell (NIH3T3) proliferation by providing better adhesiveness, biocompatibility and cytocompatibility. The antibacterial property of Zn-Ws evaluation by minimum inhibitory concentration (MIC) and zone of inhibition (ZOI) methods against clinical isolates of Gram +Ve and Gram -Ve bacterial strains have confirmed that the addition of Zn has diminished the bacterial growth and also helped in degrading the bacterial biofilms. Thus it is summed up that the process of wound healing is expected to occur with reduced risk of post-injury infections by the presence of zinc-doping on wollastonite for skin tissue application.
Collapse
Affiliation(s)
- K Mohamed Abudhahir
- Department of Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - R Murugesan
- Department of Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - R Vijayashree
- Department of Pathology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur, India
| | - Tze-Wen Chung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei, ROC
| | - A Moorthi
- Department of Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
32
|
Evaluation of Dental Pulp Stem Cell Heterogeneity and Behaviour in 3D Type I Collagen Gels. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3034727. [PMID: 32964026 PMCID: PMC7501571 DOI: 10.1155/2020/3034727] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 01/09/2023]
Abstract
Dental pulp stem cells (DPSCs) are increasingly being advocated for regenerative medicine-based therapies. However, significant heterogeneity in the genotypic/phenotypic properties of DPSC subpopulations exist, influencing their therapeutic potentials. As most studies have established DPSC heterogeneity using 2D culture approaches, we investigated whether heterogeneous DPSC proliferative and contraction/remodelling capabilities were further evident within 3D type I collagen gels in vitro. DPSC subpopulations were isolated from human third molars and identified as high/low proliferative and multipotent/unipotent, following in vitro culture expansion and population doubling (PD) analysis. High proliferative/multipotent DPSCs, such as A3 (30 PDs and 80 PDs), and low proliferative/unipotent DPSCs, such as A1 (17 PDs), were cultured in collagen gels for 12 days, either attached or detached from the surrounding culture plastic. Collagen architecture and high proliferative/multipotent DPSC morphologies were visualised by Scanning Electron Microscopy and FITC-phalloidin/Fluorescence Microscopy. DPSC proliferation (cell counts), contraction (% diameter reductions), and remodelling (MMP-2/MMP-9 gelatin zymography) of collagen gels were also evaluated. Unexpectedly, no proliferation differences existed between DPSCs, A3 (30 PDs) and A1 (17 PDs), although A3 (80 PDs) responses were significantly reduced. Despite rapid detached collagen gel contraction with A3 (30 PDs), similar contraction rates were determined with A1 (17 PDs), although A3 (80 PDs) contraction was significantly impaired. Gel contraction correlated to distinct gelatinase profiles. A3 (30 PDs) possessed superior MMP-9 and comparable MMP-2 activities to A1 (17 PDs), whereas A3 (80 PDs) had significantly reduced MMP-2/MMP-9. High proliferative/multipotent DPSCs, A3 (30 PDs), further exhibited fibroblast-like morphologies becoming polygonal within attached gels, whilst losing cytoskeletal organization and fibroblastic morphologies in detached gels. This study demonstrates that heterogeneity exists in the gel contraction and MMP expression/activity capabilities of DPSCs, potentially reflecting differences in their abilities to degrade biomaterial scaffolds and regulate cellular functions in 3D environments and their regenerative properties overall. Thus, such findings enhance our understanding of the molecular and phenotypic characteristics associated with high proliferative/multipotent DPSCs.
Collapse
|
33
|
Sun Y, Liu X, George MN, Park S, Gaihre B, Terzic A, Lu L. Enhanced nerve cell proliferation and differentiation on electrically conductive scaffolds embedded with graphene and carbon nanotubes. J Biomed Mater Res A 2020; 109:193-206. [PMID: 32441388 DOI: 10.1002/jbm.a.37016] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 04/12/2020] [Accepted: 04/19/2020] [Indexed: 12/18/2022]
Abstract
Conduits that promote nerve regeneration are currently of great medical concern, particularly when gaps exist between nerve endings. To address this issue, our laboratory previously developed a nerve conduit from biodegradable poly(caprolactone fumarate) (PCLF) that supports peripheral nerve regeneration. The present study improves upon this work by further developing an electrically conductive, positively charged PCLF scaffold through the incorporation of graphene, carbon nanotubes (CNTs), and [2-(methacryloyloxy)ethyl]trimethylammonium chloride (MTAC) (PCLF-Graphene-CNT-MTAC) using ultraviolet (UV) induced photocrosslinking. Scanning electron microscopy, transmission electron microscopy, and atomic force microscopy were used to assess the incorporation of CNTs and graphene into PCLF-Graphene-CNT-MTAC scaffolds, which displayed enhanced surface roughness and reduced electrochemical impedance when compared to neat PCLF. Scaffolds with these surface modifications also showed improved growth and differentiation of rat pheochromocytoma 12 cells in vitro, with enhanced cell growth, neurite extension, and cellular migration. Furthermore, an increased number of neurite protrusions were observed when the conduit was electrically stimulated. These results show that the electrically conductive PCLF-Graphene-CNT-MTAC nerve scaffolds presented here support the cellular behaviors that are critical for nerve regeneration, ultimately making this material an attractive candidate for regenerative medicine applications.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew N George
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Sungjo Park
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Andre Terzic
- Department of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
34
|
You P, Liu Y, Wang X, Li B, Wu W, Tang L. Acellular pericardium: A naturally hierarchical, osteoconductive, and osteoinductive biomaterial for guided bone regeneration. J Biomed Mater Res A 2020; 109:132-145. [PMID: 32441432 DOI: 10.1002/jbm.a.37011] [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] [Received: 11/17/2019] [Revised: 04/15/2020] [Accepted: 04/19/2020] [Indexed: 01/04/2023]
Abstract
There is great demand for an improved barrier membrane with osteogenic potential for guided bone regeneration (GBR). Natural acellular porcine pericardium (APP) is increasingly used in regenerative medicine as a kind of common extracellular matrix materials. This study aimed to investigate its potential application in GBR, especially its osteoconductive and osteoinductive properties. Bio-Gide (BG), a commercial collagen membrane, was set as the control group. APP samples were characterized by physicochemical analyses and their biological effects on human bone mesenchymal stem cells (hBMSCs) and human gingival fibroblasts (hGFs) were also examined. Additionally, the osteogenic potential of APP was tested on a bilateral critical-sized calvarial defect model. We discovered that the smooth surface of APP tended to recruit more hBMSCs. Moreover, promoted proliferation and osteogenic differentiation of hBMSCs was detected on this side of APP, with increased alkaline phosphatase activity and upregulated expression of bone-specific genes. Besides, the rough side of APP showed good biocompatibility and barrier function with hGFs. Histologic observation and analysis of calvarial defect healing over 4 weeks revealed enhanced bone regeneration under APP compared with BG and the control group. The results of this study indicate that APP is a potential osteoconductive and osteoinductive biomaterial for GBR.
Collapse
Affiliation(s)
- Pengyue You
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, No.22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, P.R. China
| | - Yuhua Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, No.22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, P.R. China
| | - Xinzhi Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, No.22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, P.R. China
| | - Bowen Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, No.22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, P.R. China
| | - Weiyi Wu
- Department of Second Clinical Division, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, No.22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, P.R. China
| | - Lin Tang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, No.22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, P.R. China
| |
Collapse
|
35
|
Zhao H, Tang J, Zhou D, Weng Y, Qin W, Liu C, Lv S, Wang W, Zhao X. Electrospun Icariin-Loaded Core-Shell Collagen, Polycaprolactone, Hydroxyapatite Composite Scaffolds for the Repair of Rabbit Tibia Bone Defects. Int J Nanomedicine 2020; 15:3039-3056. [PMID: 32431500 PMCID: PMC7200251 DOI: 10.2147/ijn.s238800] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Background Electrospinning is a widely used technology that can produce scaffolds with high porosity and surface area for bone regeneration. However, the small pore sizes in electrospun scaffolds constrain cell growth and tissue-ingrowth. In this study, novel drug-loading core-shell scaffolds were fabricated via electrospinning and freeze drying to facilitate the repair of tibia bone defects in rabbit models. Materials and Methods The collagen core scaffolds were freeze-dried containing icariin (ICA)-loaded chitosan microspheres. The shell scaffolds were electrospun using collagen, polycaprolactone and hydroxyapatite materials to form CPH composite scaffolds with the ones containing ICA microspheres named CPHI. The core-shell scaffolds were then cross-linked by genipin. The morphology, microstructure, physical and mechanical properties of the scaffolds were assessed. Rat marrow mesenchymal stem cells from the wistar rat were cultured with the scaffolds. The cell adhesion and proliferation were analysed. Adult rabbit models with tibial plateau defects were used to evaluate the performance of these scaffolds in repairing the bone defects over 4 to 12 weeks. Results The results reveal that the novel drug-loading core-shell scaffolds were successfully fabricated, which showed good physical and chemical properties and appropriate mechanical properties. Furthermore, excellent cells attachment was observed on the CPHI scaffolds. The results from radiography, micro-computed tomography, histological and immunohistochemical analysis demonstrated that abundant new bones were formed on the CPHI scaffolds. Conclusion These new core-shell composite scaffolds have great potential for bone tissue engineering applications and may lead to effective bone regeneration and repair.
Collapse
Affiliation(s)
- Hongbin Zhao
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Junjie Tang
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Dong Zhou
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Yiping Weng
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Wen Qin
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Chun Liu
- Medical Research Centre, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213164, People's Republic of China
| | - Songwei Lv
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China
| | - Wei Wang
- Medical School, Hexi University, Zhangye 730041, People's Republic of China
| | - Xiubo Zhao
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, People's Republic of China.,Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
36
|
Begum R, Perriman AW, Su B, Scarpa F, Kafienah W. Chondroinduction of Mesenchymal Stem Cells on Cellulose-Silk Composite Nanofibrous Substrates: The Role of Substrate Elasticity. Front Bioeng Biotechnol 2020; 8:197. [PMID: 32266231 PMCID: PMC7096586 DOI: 10.3389/fbioe.2020.00197] [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: 09/29/2019] [Accepted: 02/28/2020] [Indexed: 01/09/2023] Open
Abstract
Smart biomaterials with an inherent capacity to elicit specific behaviors in lieu of biological prompts would be advantageous for regenerative medicine applications. In this work, we employ an electrospinning technique to model the in vivo nanofibrous extracellular matrix (ECM) of cartilage using a chondroinductive cellulose and silk polymer blend (75:25 ratio). This natural polymer composite is directly electrospun for the first time, into nanofibers without post-spun treatment, using a trifluoroacetic acid and acetic acid cosolvent system. Biocompatibility of the composite nanofibres with human mesenchymal stem cells (hMSCs) is demonstrated and its inherent capacity to direct chondrogenic stem cell differentiation, in the absence of stimulating growth factors, is confirmed. This chondrogenic stimulation could be countered biochemically using fibroblast growth factor-2, a growth factor used to enhance the proliferation of hMSCs. Furthermore, the potential mechanisms driving this chondroinduction at the cell-biomaterial interface is investigated. Composite substrates are fabricated as two-dimensional film surfaces and cultured with hMSCs in the presence of chemicals that interfere with their biochemical and mechanical signaling pathways. Preventing substrate surface elasticity transmission resulted in a significant downregulation of chondrogenic gene expression. Interference with the classical chondrogenic Smad2/3 phosphorylation pathway did not impact chondrogenesis. The results highlight the importance of substrate mechanical elasticity on hMSCs chondroinduction and its independence to known chondrogenic biochemical pathways. The newly fabricated scaffolds provide the foundation for designing a robust, self-inductive, and cost-effective biomimetic biomaterial for cartilage tissue engineering.
Collapse
Affiliation(s)
- Runa Begum
- Faculty of Biomedical Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Adam W Perriman
- Faculty of Biomedical Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Bo Su
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Fabrizio Scarpa
- Bristol Composites Institute (ACCIS), University of Bristol, Bristol, United Kingdom
| | - Wael Kafienah
- Faculty of Biomedical Sciences, School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
37
|
Ehlert M, Radtke A, Jędrzejewski T, Roszek K, Bartmański M, Piszczek P. In Vitro Studies on Nanoporous, Nanotubular and Nanosponge-Like Titania Coatings, with the Use of Adipose-Derived Stem Cells. MATERIALS 2020; 13:ma13071574. [PMID: 32235354 PMCID: PMC7177883 DOI: 10.3390/ma13071574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022]
Abstract
In vitro biological research on a group of amorphous titania coatings of different nanoarchitectures (nanoporous, nanotubular, and nanosponge-like) produced on the surface of Ti6Al4V alloy samples have been carried out, aimed at assessing their ability to interact with adipose-derived mesenchymal stem cells (ADSCs) and affect their activity. The attention has been drawn to the influence of surface coating architecture and its physicochemical properties on the ADSCs proliferation. Moreover, in vitro co-cultures: (1) fibroblasts cell line L929/ADSCs and (2) osteoblasts cell line MG-63/ADSCs on nanoporous, nanotubular and nanosponge-like TiO2 coatings have been studied. This allowed for evaluating the impact of the surface properties, especially roughness and wettability, on the creation of the beneficial microenvironment for co-cultures and/or enhancing differentiation potential of stem cells. Obtained results showed that the nanoporous surface is favorable for ADSCs, has great biointegrative properties, and supports the growth of co-cultures with MG-63 osteoblasts and L929 fibroblasts. Additionally, the number of osteoblasts seeded and cultured with ADSCs on TNT5 surface raised after 72-h culture almost twice when compared with the unmodified scaffold and by 30% when compared with MG-63 cells growing alone. The alkaline phosphatase activity of MG-63 osteoblasts co-cultured with ADSCs increased, that indirectly confirmed our assumptions that TNT-modified scaffolds create the osteogenic niche and enhance osteogenic potential of ADSCs.
Collapse
Affiliation(s)
- Michalina Ehlert
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-implant Ltd. Gagarina 5/102, 87-100 Toruń, Poland
| | - Aleksandra Radtke
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-implant Ltd. Gagarina 5/102, 87-100 Toruń, Poland
- Correspondence: (A.R.); (P.P.); Tel.: +48-600321294 (A.R.); Tel.: +48-607883357 (P.P.)
| | - Tomasz Jędrzejewski
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Katarzyna Roszek
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Michał Bartmański
- Faculty of Mechanical Engineering, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland;
| | - Piotr Piszczek
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-implant Ltd. Gagarina 5/102, 87-100 Toruń, Poland
- Correspondence: (A.R.); (P.P.); Tel.: +48-600321294 (A.R.); Tel.: +48-607883357 (P.P.)
| |
Collapse
|
38
|
Swetha S, Lavanya K, Sruthi R, Selvamurugan N. An insight into cell-laden 3D-printed constructs for bone tissue engineering. J Mater Chem B 2020; 8:9836-9862. [DOI: 10.1039/d0tb02019b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, we have spotlighted various combinations of bioinks to optimize the biofabrication of 3D bone constructs.
Collapse
Affiliation(s)
- S. Swetha
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - K. Lavanya
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - R. Sruthi
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - N. Selvamurugan
- Department of Biotechnology, College of Engineering and Technology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| |
Collapse
|
39
|
Chimpibul W, Nakaji-Hirabayashi T, Yuan X, Matsumura K. Controlling the degradation of cellulose scaffolds with Malaprade oxidation for tissue engineering. J Mater Chem B 2020; 8:7904-7913. [DOI: 10.1039/d0tb01015d] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cellulose scaffolds, whose biodegradation can be controlled through the reaction with amine compounds in the human body, were developed for tissue engineering applications.
Collapse
Affiliation(s)
| | | | - Xida Yuan
- School of Materials Science
- Japan Advanced Institute of Science and Technology
- Ishikawa
- Japan
| | - Kazuaki Matsumura
- School of Materials Science
- Japan Advanced Institute of Science and Technology
- Ishikawa
- Japan
| |
Collapse
|
40
|
Melica ME, La Regina G, Parri M, Peired AJ, Romagnani P, Lasagni L. Substrate Stiffness Modulates Renal Progenitor Cell Properties via a ROCK-Mediated Mechanotransduction Mechanism. Cells 2019; 8:cells8121561. [PMID: 31816967 PMCID: PMC6953094 DOI: 10.3390/cells8121561] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 01/11/2023] Open
Abstract
Stem cell (SC)-based tissue engineering and regenerative medicine (RM) approaches may provide alternative therapeutic strategies for the rising number of patients suffering from chronic kidney disease. Embryonic SCs and inducible pluripotent SCs are the most frequently used cell types, but autologous patient-derived renal SCs, such as human CD133+CD24+ renal progenitor cells (RPCs), represent a preferable option. RPCs are of interest also for the RM approaches based on the pharmacological encouragement of in situ regeneration by endogenous SCs. An understanding of the biochemical and biophysical factors that influence RPC behavior is essential for improving their applicability. We investigated how the mechanical properties of the substrate modulate RPC behavior in vitro. We employed collagen I-coated hydrogels with variable stiffness to modulate the mechanical environment of RPCs and found that their morphology, proliferation, migration, and differentiation toward the podocyte lineage were highly dependent on mechanical stiffness. Indeed, a stiff matrix induced cell spreading and focal adhesion assembly trough a Rho kinase (ROCK)-mediated mechanism. Similarly, the proliferative and migratory capacity of RPCs increased as stiffness increased and ROCK inhibition, by either Y27632 or antisense LNA-GapmeRs, abolished these effects. The acquisition of podocyte markers was also modulated, in a narrow range, by the elastic modulus and involved ROCK activity. Our findings may aid in 1) the optimization of RPC culture conditions to favor cell expansion or to induce efficient differentiation with important implication for RPC bioprocessing, and in 2) understanding how alterations of the physical properties of the renal tissue associated with diseases could influenced the regenerative response of RPCs.
Collapse
Affiliation(s)
- Maria Elena Melica
- Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Viale Morgagni 50, 50136 Florence, Italy; (M.E.M.); (A.J.P.); (P.R.)
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (G.L.R.); (M.P.)
| | - Gilda La Regina
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (G.L.R.); (M.P.)
| | - Matteo Parri
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (G.L.R.); (M.P.)
| | - Anna Julie Peired
- Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Viale Morgagni 50, 50136 Florence, Italy; (M.E.M.); (A.J.P.); (P.R.)
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (G.L.R.); (M.P.)
| | - Paola Romagnani
- Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Viale Morgagni 50, 50136 Florence, Italy; (M.E.M.); (A.J.P.); (P.R.)
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (G.L.R.); (M.P.)
- Nephrology Unit and Meyer Children’s University Hospital, Viale Pieraccini 24, 50139 Florence, Italy
| | - Laura Lasagni
- Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Viale Morgagni 50, 50136 Florence, Italy; (M.E.M.); (A.J.P.); (P.R.)
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (G.L.R.); (M.P.)
- Correspondence: ; Tel.: +39-055-2758165
| |
Collapse
|
41
|
Xu Y, Chen C, Hellwarth PB, Bao X. Biomaterials for stem cell engineering and biomanufacturing. Bioact Mater 2019; 4:366-379. [PMID: 31872161 PMCID: PMC6909203 DOI: 10.1016/j.bioactmat.2019.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/09/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Recent years have witnessed the expansion of tissue failures and diseases. The uprising of regenerative medicine converges the sight onto stem cell-biomaterial based therapy. Tissue engineering and regenerative medicine proposes the strategy of constructing spatially, mechanically, chemically and biologically designed biomaterials for stem cells to grow and differentiate. Therefore, this paper summarized the basic properties of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells. The properties of frequently used biomaterials were also described in terms of natural and synthetic origins. Particularly, the combination of stem cells and biomaterials for tissue repair applications was reviewed in terms of nervous, cardiovascular, pancreatic, hematopoietic and musculoskeletal system. Finally, stem-cell-related biomanufacturing was envisioned and the novel biofabrication technologies were discussed, enlightening a promising route for the future advancement of large-scale stem cell-biomaterial based therapeutic manufacturing.
Collapse
Affiliation(s)
| | | | | | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, West Lafayette, IN, 47907, USA
| |
Collapse
|
42
|
Sun J, Ma X, Chu HT, Feng B, Tuan RS, Jiang Y. Biomaterials and Advanced Biofabrication Techniques in hiPSCs Based Neuromyopathic Disease Modeling. Front Bioeng Biotechnol 2019; 7:373. [PMID: 31850331 PMCID: PMC6895005 DOI: 10.3389/fbioe.2019.00373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are reprogrammed somatic cells by defined factors, and have great application potentials in tissue regeneration and disease modeling. Biomaterials have been widely used in stem cell-based studies, and are involved in human iPSCs based studies, but they were not enough emphasized and recognized. Biomaterials can mimic the extracellular matrix and microenvironment, and act as powerful tools to promote iPSCs proliferation, differentiation, maturation, and migration. Many classic and advanced biofabrication technologies, such as cell-sheet approach, electrospinning, and 3D-bioprinting, are used to provide physical cues in macro-/micro-patterning, and in combination with other biological factors to support iPSCs applications. In this review, we highlight the biomaterials and fabrication technologies used in human iPSC-based tissue engineering to model neuromyopathic diseases, particularly those with genetic mutations, such as Duchenne Muscular Dystrophy (DMD), Congenital Heart Diseases (CHD) and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Jing Sun
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xun Ma
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ho Ting Chu
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bo Feng
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Rocky S Tuan
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yangzi Jiang
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
43
|
Ehlert M, Roszek K, Jędrzejewski T, Bartmański M, Radtke A. Titania Nanofiber Scaffolds with Enhanced Biointegration Activity-Preliminary In Vitro Studies. Int J Mol Sci 2019; 20:E5642. [PMID: 31718064 PMCID: PMC6888681 DOI: 10.3390/ijms20225642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 12/11/2022] Open
Abstract
The increasing need for novel bone replacement materials has been driving numerous studies on modifying their surface to stimulate osteogenic cells expansion and to accelerate bone tissue regeneration. The goal of the presented study was to optimize the production of titania-based bioactive materials with high porosity and defined nanostructure, which supports the cell viability and growth. We have chosen to our experiments TiO2 nanofibers, produced by chemical oxidation of Ti6Al4V alloy. Fibrous nanocoatings were characterized structurally (X-ray diffraction (XRD)) and morphologically (scanning electron microscopy (SEM)). The wettability of the coatings and their mechanical properties were also evaluated. We have investigated the direct influence of the modified titanium alloy surfaces on the survival and proliferation of mesenchymal stem cells derived from adipose tissue (ADSCs). In parallel, proliferation of bone tissue cells-human osteoblasts MG-63 and connective tissue cells - mouse fibroblasts L929, as well as cell viability in co-cultures (osteoblasts/ADSCs and fibroblasts/ADSCs has been studied. The results of our experiments proved that among all tested nanofibrous coatings, the amorphous titania-based ones were the most optimal scaffolds for the integration and proliferation of ADSCs, fibroblasts, and osteoblasts. Thus, we postulated these scaffolds to have the osteopromotional potential. However, from the co-culture experiments it can be concluded that ADSCs have the ability to functionalize the initially unfavorable surface, and make it suitable for more specialized and demanding cells.
Collapse
Affiliation(s)
- Michalina Ehlert
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-Implant Ltd., Gagarina 5/102, 87-100 Toruń, Poland
| | - Katarzyna Roszek
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Tomasz Jędrzejewski
- Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland; (K.R.); (T.J.)
| | - Michał Bartmański
- Faculty of Mechanical Engineering, Gdańsk University of Technology, Gabriela Narutowicza 11/12, 80-233 Gdańsk, Poland;
| | - Aleksandra Radtke
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100 Toruń, Poland;
- Nano-Implant Ltd., Gagarina 5/102, 87-100 Toruń, Poland
| |
Collapse
|
44
|
Narayanamurthy V, Samsuri F, Firus Khan AY, Hamzah HA, Baharom MB, Kumary TV, Anil Kumar PR, Raj DK. Direct cell imprint lithography in superconductive carbon black polymer composites: process optimization, characterization and in vitro toxicity analysis. BIOINSPIRATION & BIOMIMETICS 2019; 15:016002. [PMID: 30897554 DOI: 10.1088/1748-3190/ab1243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cell imprint lithography (CIL) or cell replication plays a vital role in fields like biomimetic smart culture substrates, bone tissue engineering, cell guiding, cell adhesion, tissue engineering, cell microenvironments, tissue microenvironments, cell research, drug delivery, diagnostics, therapeutics and many other applications. Herein we report a new formulation of superconductive carbon black photopolymer composite and its characterization towards a CIL process technique. In this article, we demonstrated an approach of using a carbon nanoparticle-polymer composite (CPC) for patterning cells. It is observed that a 0.3 wt % load of carbon nanoparticles (CNPs) in a carbon polymer mixture (CPM) was optimal for cell-imprint replica fabrication. The electrical resistance of the 3-CPC (0.3 wt %) was reduced by 68% when compared to N-CPC (0 wt %). This method successfully replicated the single cell with sub-organelle scale. The shape of microvesicles, grooves, pores, blebs or microvilli on the cellular surface was patterned clearly. This technique delivers a free-standing cell feature substrate. In vitro evaluation of the polymer demonstrated it as an ideal candidate for biomimetic biomaterial applications. This approach also finds its application in study based on morphology, especially for drug delivery applications and for investigations based on molecular pathways.
Collapse
Affiliation(s)
- Vigneswaran Narayanamurthy
- Faculty of Electrical and Electronics Engineering, University Malaysia Pahang, Pekan 26600, Malaysia. Faculty of Medicine, International Islamic University Malaysia, Kuantan, Pahang 25200, Malaysia
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Argentati C, Morena F, Tortorella I, Bazzucchi M, Porcellati S, Emiliani C, Martino S. Insight into Mechanobiology: How Stem Cells Feel Mechanical Forces and Orchestrate Biological Functions. Int J Mol Sci 2019; 20:E5337. [PMID: 31717803 PMCID: PMC6862138 DOI: 10.3390/ijms20215337] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
The cross-talk between stem cells and their microenvironment has been shown to have a direct impact on stem cells' decisions about proliferation, growth, migration, and differentiation. It is well known that stem cells, tissues, organs, and whole organisms change their internal architecture and composition in response to external physical stimuli, thanks to cells' ability to sense mechanical signals and elicit selected biological functions. Likewise, stem cells play an active role in governing the composition and the architecture of their microenvironment. Is now being documented that, thanks to this dynamic relationship, stemness identity and stem cell functions are maintained. In this work, we review the current knowledge in mechanobiology on stem cells. We start with the description of theoretical basis of mechanobiology, continue with the effects of mechanical cues on stem cells, development, pathology, and regenerative medicine, and emphasize the contribution in the field of the development of ex-vivo mechanobiology modelling and computational tools, which allow for evaluating the role of forces on stem cell biology.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Serena Porcellati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (F.M.); (I.T.); (M.B.); (S.P.); (C.E.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| |
Collapse
|
46
|
Rodrigues C, Naasani LIS, Zanatelli C, Paim TC, Azevedo JG, de Lima JC, da Cruz Fernandes M, Buchner S, Wink MR. Bioglass 45S5: Structural characterization of short range order and analysis of biocompatibility with adipose-derived mesenchymal stromal cells in vitro and in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109781. [DOI: 10.1016/j.msec.2019.109781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 05/12/2019] [Accepted: 05/20/2019] [Indexed: 12/11/2022]
|
47
|
Gryshkov O, Müller M, Leal-Marin S, Mutsenko V, Suresh S, Kapralova VM, Glasmacher B. Advances in the application of electrohydrodynamic fabrication for tissue engineering. ACTA ACUST UNITED AC 2019. [DOI: 10.1088/1742-6596/1236/1/012024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
48
|
Valencia Zapata ME, Mina Hernandez JH, Grande Tovar CD, Valencia Llano CH, Diaz Escobar JA, Vázquez-Lasa B, San Román J, Rojo L. Novel Bioactive and Antibacterial Acrylic Bone Cement Nanocomposites Modified with Graphene Oxide and Chitosan. Int J Mol Sci 2019; 20:ijms20122938. [PMID: 31208091 PMCID: PMC6627441 DOI: 10.3390/ijms20122938] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/09/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Acrylic bone cements (ABCs) have played a key role in orthopedic surgery mainly in arthroplasties, but their use is increasingly extending to other applications, such as remodeling of cancerous bones, cranioplasties, and vertebroplasties. However, these materials present some limitations related to their inert behavior and the risk of infection after implantation, which leads to a lack of attachment and makes necessary new surgical interventions. In this research, the physicochemical, thermal, mechanical, and biological properties of ABCs modified with chitosan (CS) and graphene oxide (GO) were studied. Fourier transform infrared (FTIR) spectroscopy, proton nuclear magnetic resonance (1H-NMR) scanning electron microscopy (SEM), Raman mapping, thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), compression resistance, mechanical dynamic analysis (DMA), hydrolytic degradation, cell viability, alkaline phosphatase (ALP) activity with human osteoblasts (HOb), and antibacterial activity against Gram-negative bacteria Escherichia coli were used to characterize the ABCs. The results revealed good dispersion of GO nanosheets in the ABCs. GO provided an increase in antibacterial activity, roughness, and flexural behavior, while CS generated porosity, increased the rate of degradation, and decreased compression properties. All ABCs were not cytotoxic and support good cell viability of HOb. The novel formulation of ABCs containing GO and CS simultaneously, increased the thermal stability, flexural modulus, antibacterial behavior, and osteogenic activity, which gives it a high potential for its uses in orthopedic applications.
Collapse
Affiliation(s)
- Mayra Eliana Valencia Zapata
- Grupo de Materiales Compuestos, Escuela de Ingeniería de Materiales, Universidad del Valle, Calle 13 # 100-00, Cali 76001, Colombia.
| | - José Herminsul Mina Hernandez
- Grupo de Materiales Compuestos, Escuela de Ingeniería de Materiales, Universidad del Valle, Calle 13 # 100-00, Cali 76001, Colombia.
| | - Carlos David Grande Tovar
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia.
| | | | - José Alfredo Diaz Escobar
- Departamento de Ciencias Básicas, Institución Universitaria Antonio José Camacho, Avenida 6N # 28N - 102, Cali 76001, Colombia.
| | - Blanca Vázquez-Lasa
- Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain.
- Consorcio Centro de Investigación Biomedica en red, CIBER-BBN, 28029 Madrid, Spain.
| | - Julio San Román
- Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain.
- Consorcio Centro de Investigación Biomedica en red, CIBER-BBN, 28029 Madrid, Spain.
| | - Luis Rojo
- Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva 3, 28006 Madrid, Spain.
- Consorcio Centro de Investigación Biomedica en red, CIBER-BBN, 28029 Madrid, Spain.
| |
Collapse
|
49
|
Mestres G, Perez RA, D’Elía NL, Barbe L. Advantages of microfluidic systems for studying cell-biomaterial interactions—focus on bone regeneration applications. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab1033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
50
|
Voga M, Drnovsek N, Novak S, Majdic G. Silk fibroin induces chondrogenic differentiation of canine adipose-derived multipotent mesenchymal stromal cells/mesenchymal stem cells. J Tissue Eng 2019; 10:2041731419835056. [PMID: 30899447 PMCID: PMC6419250 DOI: 10.1177/2041731419835056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/11/2019] [Indexed: 12/19/2022] Open
Abstract
Under appropriate culture conditions, mesenchymal stem cells (MSC), also called more properly multipotent mesenchymal stromal cells (MMSC), can be induced toward differentiation into different cell lineages. In order to guide stem cell fate within an environment resembling the stem cell niche, different biomaterials are being developed. In the present study, we used silk fibroin (SF) as a biomaterial supporting the growth of MMSC and studied its effect on chondrogenesis of canine adipose–derived MMSC (cADMMSC). Adipose tissue was collected from nine privately owned dogs. MMSC were cultured on SF films and SF scaffolds in a standard cell culture medium. Cell morphology was evaluated by scanning electron microscopy (SEM). Chondrogenic differentiation was evaluated by alcian blue staining and mRNA expression of collagen type 1, collagen type 2, Sox9, and Aggrecan genes. cADMMSC cultured on SF films and SF scaffolds stained positive using alcian blue. SEM images revealed nodule-like structures with matrix vesicles and fibers resembling chondrogenic nodules. Gene expression of chondrogenic markers Sox9 and Aggrecan were statistically significantly upregulated in cADMMSC cultured on SF films in comparison to negative control cADMMSC. This result suggests that chondrogenesis of cADMMSC could occur when cells were grown on SF films in a standard cell culture medium without specific culture conditions, which were previously considered necessary for induction of chondrogenic differentiation.
Collapse
Affiliation(s)
- Metka Voga
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Natasa Drnovsek
- Department for Nanostructured Materials, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Sasa Novak
- Department for Nanostructured Materials, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Gregor Majdic
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia.,Institute of Physiology, Medical School, University of Maribor, Maribor, Slovenia
| |
Collapse
|