1
|
Nie Y, Mu Q, Sun Y, Ferdous Z, Wang L, Chen C, Nakajima T, Gong JP, Tanaka S, Tsuda M. Mechanochemistry-Induced Universal Hydrogel Surface Modification for Orientation and Enhanced Differentiation of Skeletal Muscle Myoblasts. ACS APPLIED BIO MATERIALS 2025; 8:3144-3155. [PMID: 40106521 DOI: 10.1021/acsabm.4c01991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Micropatterned surface substrates containing topographic cues offer the possibility of programming tissue organization as a cell template by guiding cell alignment, adhesion, and function. In this study, we developed and used a force stamp method to grow aligned micropatterns with tunable chemical properties and elasticity on the surface of hydrogels based on a force-triggered polymerization mechanism of double-network hydrogels to elucidate the underlying mechanisms by which cells sense and respond to their mechanical and chemical microenvironments. In this work, we describe the impact of aligned micropatterns on the combined effects of microstructural chemistry and mechanics on the selective adhesion, directed migration, and differentiation of myoblasts. Our investigations revealed that topographically engineered substrates with hydrophobic and elevated surface roughness significantly enhanced myoblast adhesion kinetics. Concurrently, spatially ordered architectures facilitated cytoskeletal reorganization in myocytes, establishing biomechanically favorable niches for syncytial myotube development through extracellular matrix (ECM) physical guidance. Reverse transcription PCR analysis and immunofluorescence revealed that the expression of differentiation-specific genes, myosin heavy chain, and myogenic regulatory factors Myf5 and MyoD was upregulated in muscle cells on the aligned patterned scaffolds. These results suggest that the aligned micropatterns can promote muscle cell differentiation, making them potential scaffolds for enhancing skeletal differentiation.
Collapse
Affiliation(s)
- Yuheng Nie
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Qifeng Mu
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Yanpeng Sun
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Zannatul Ferdous
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
| | - Cewen Chen
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Tasuku Nakajima
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
- Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Jian Ping Gong
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
- Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
- Department of Surgical Pathology, Hokkaido University Hospital, N14W5, Kita-ku, Sapporo 060-8648, Japan
| | - Masumi Tsuda
- Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan
| |
Collapse
|
2
|
Chen Z, Xu C, Chen X, Huang J, Guo Z. Advances in Electrically Conductive Hydrogels: Performance and Applications. SMALL METHODS 2025; 9:e2401156. [PMID: 39529563 DOI: 10.1002/smtd.202401156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Electrically conductive hydrogels are highly hydrated 3D networks consisting of a hydrophilic polymer skeleton and electrically conductive materials. Conductive hydrogels have excellent mechanical and electrical properties and have further extensive application prospects in biomedical treatment and other fields. Whereas numerous electrically conductive hydrogels have been fabricated, a set of general principles, that can rationally guide the synthesis of conductive hydrogels using different substances and fabrication methods for various application scenarios, remain a central demand of electrically conductive hydrogels. This paper systematically summarizes the processing, performances, and applications of conductive hydrogels, and discusses the challenges and opportunities in this field. In view of the shortcomings of conductive hydrogels in high electrical conductivity, matchable mechanical properties, as well as integrated devices and machines, it is proposed to synergistically design and process conductive hydrogels with applications in complex surroundings. It is believed that this will present a fresh perspective for the research and development of conductive hydrogels, and further expand the application of conductive hydrogels.
Collapse
Affiliation(s)
- Zhiwei Chen
- Ministry of Education Key Laboratory for the Green Preparation and Applications, Hubei University, Wuhan, 430062, China
| | - Chenggong Xu
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xionggang Chen
- Ministry of Education Key Laboratory for the Green Preparation and Applications, Hubei University, Wuhan, 430062, China
| | - Jinxia Huang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Zhiguang Guo
- Ministry of Education Key Laboratory for the Green Preparation and Applications, Hubei University, Wuhan, 430062, China
| |
Collapse
|
3
|
Dhoundiyal S, Sharma A, Alam MA. Fiber Technology in Drug Delivery and Pharmaceuticals. Curr Drug Deliv 2025; 22:261-282. [PMID: 38279740 DOI: 10.2174/0115672018279628231221105210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 01/28/2024]
Abstract
The field of fiber technology is a dynamic and innovative domain that offers novel solutions for controlled and targeted therapeutic interventions. This abstract provides an overview of key aspects within this field, encompassing a range of techniques, applications, commercial developments, intellectual property, and regulatory considerations. The foundational introduction establishes the significance of fiber-based drug delivery systems. Electrospinning, a pivotal technique, has been explored in this paper, along with its various methods and applications. Monoaxial, coaxial, triaxial, and side-by-side electrospinning techniques each offer distinct advantages and applications. Centrifugal spinning, solution and melt blowing spinning, and pressurized gyration further contribute to the field's diversity. The review also delves into commercial advancements, highlighting marketed products that have successfully harnessed fiber technology. The role of intellectual property is acknowledged, with patents reflecting the innovative strides in fiber-based drug delivery. The regulatory perspective, essential for ensuring safety and efficacy, is discussed in the context of global regulatory agencies' evaluations. This review encapsulates the multidimensional nature of fiber technology in drug delivery and pharmaceuticals, showcasing its potential to revolutionize medical treatments and underscores the importance of continued collaboration between researchers, industry, and regulators for its advancement.
Collapse
Affiliation(s)
- Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Aditya Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
4
|
Ghosh S, Bhatti GK, Sharma PK, Kandimalla R, Mastana SS, Bhatti JS. Potential of Nano-Engineered Stem Cells in the Treatment of Multiple Sclerosis: A Comprehensive Review. Cell Mol Neurobiol 2023; 44:6. [PMID: 38104307 PMCID: PMC11397842 DOI: 10.1007/s10571-023-01434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023]
Abstract
Multiple sclerosis (MS) is a chronic and degrading autoimmune disorder mainly targeting the central nervous system, leading to progressive neurodegeneration, demyelination, and axonal damage. Current treatment options for MS are limited in efficacy, generally linked to adverse side effects, and do not offer a cure. Stem cell therapies have emerged as a promising therapeutic strategy for MS, potentially promoting remyelination, exerting immunomodulatory effects and protecting against neurodegeneration. Therefore, this review article focussed on the potential of nano-engineering in stem cells as a therapeutic approach for MS, focusing on the synergistic effects of combining stem cell biology with nanotechnology to stimulate the proliferation of oligodendrocytes (OLs) from neural stem cells and OL precursor cells, by manipulating neural signalling pathways-PDGF, BMP, Wnt, Notch and their essential genes such as Sox, bHLH, Nkx. Here we discuss the pathophysiology of MS, the use of various types of stem cells in MS treatment and their mechanisms of action. In the context of nanotechnology, we present an overview of its applications in the medical and research field and discuss different methods and materials used to nano-engineer stem cells, including surface modification, biomaterials and scaffolds, and nanoparticle-based delivery systems. We further elaborate on nano-engineered stem cell techniques, such as nano script, nano-exosome hybrid, nano-topography and their potentials in MS. The article also highlights enhanced homing, engraftment, and survival of nano-engineered stem cells, targeted and controlled release of therapeutic agents, and immunomodulatory and tissue repair effects with their challenges and limitations. This visual illustration depicts the process of utilizing nano-engineering in stem cells and exosomes for the purpose of delivering more accurate and improved treatments for Multiple Sclerosis (MS). This approach targets specifically the creation of oligodendrocytes, the breakdown of which is the primary pathological factor in MS.
Collapse
Affiliation(s)
- Sushruta Ghosh
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University, Rajasthan, India
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University, Rajasthan, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana, India
- Department of Applied Biology, CSIR-Indian Institute of Technology, Hyderabad, India
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India.
| |
Collapse
|
5
|
Doğan D, Karaduman FR, Horzum N, Metin AÜ. Boron nitride decorated poly(vinyl alcohol)/poly(acrylic acid) composite nanofibers: A promising material for biomedical applications. J Mech Behav Biomed Mater 2023; 141:105773. [PMID: 36934687 DOI: 10.1016/j.jmbbm.2023.105773] [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: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/09/2023]
Abstract
In this study, polyvinyl alcohol (PVA) and polyacrylic acid (PAA) nanofibers loaded with boron nitride nanoparticles (mBN) were fabricated by using electrospinning and crosslinked by heat treatment. The physical, chemical, and mechanical properties, hydrophilic behavior, and degradability of composite nanofibers were evaluated. The mechanical properties such as elastic modulus, elongation percentage at the break, and mechanical strength of PVA/PAA nanofibers improved with mBN loading. The thermal conductivity of composite nanofibers reached 0.12 W/m·K at mBN content of 1.0 wt% due to the continuous heat conduction pathways of mBN. In the meantime, while there was no cytotoxicity recorded for both L929 and HUVEC cell lines for all composite nanofibers, the antimicrobial efficiency improved with the incorporation of mBN compared with PVA/PAA and recorded as 68.8% and 75.1% for Escherichia coli and Staphylococcus aureus, respectively. On this basis, the present work proposes a promising biomaterial for biomedical applications such as dual drug delivery, particularly including both hydrophobic and hydrophilic drugs or wound dressing.
Collapse
Affiliation(s)
- Deniz Doğan
- Department of Chemistry, Faculty of Science and Arts, Kırıkkale University, Yahşihan, 71450, Kırıkkale, Turkey
| | - F Rabia Karaduman
- Graduate School of Natural and Applied Sciences, İzmir Katip Çelebi University, İzmir, 35620, Turkey
| | - Nesrin Horzum
- Department of Engineering Sciences, Izmir Katip Çelebi University, Izmir, Turkey
| | - Ayşegül Ülkü Metin
- Department of Chemistry, Faculty of Science and Arts, Kırıkkale University, Yahşihan, 71450, Kırıkkale, Turkey.
| |
Collapse
|
6
|
Chen Y, Lock J, Liu HH. Nanocomposites for cartilage regeneration. Nanomedicine (Lond) 2023. [DOI: 10.1016/b978-0-12-818627-5.00018-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
|
7
|
Bate TSR, Shanahan W, Casillo JP, Grant R, Forbes SJ, Callanan A. Rat liver ECM incorporated into electrospun polycaprolactone scaffolds as a platform for hepatocyte culture. J Biomed Mater Res B Appl Biomater 2022; 110:2612-2623. [PMID: 35734943 PMCID: PMC9796056 DOI: 10.1002/jbm.b.35115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/13/2022] [Accepted: 06/08/2022] [Indexed: 12/30/2022]
Abstract
Liver disease is expanding across the globe; however, health-care systems still lack approved pharmaceutical treatment strategies to mitigate potential liver failures. Organ transplantation is the only treatment for liver failure and with increasing cases of liver disease, transplant programs increasingly cannot provide timely transplant availability for all patients. The development of pharmaceutical mitigation strategies is clearly necessary and methods to improve drug development processes are considered vital for this purpose. Herein, we present a methodology for incorporating whole organ decellularised rat liver ECM (rLECM) into polycaprolactone (PCL) electrospun scaffolds with the aim of producing biologically relevant liver tissue models. rLECM PCL scaffolds have been produced with 5 w/w% and 10 w/w% rLECM:PCL and were analyzed by SEM imaging, tensile mechanical analyses and FTIR spectroscopy. The hepatocellular carcinoma cell line, HepG2, was cultured upon the scaffolds for 14 days and were analyzed through cell viability assay, DNA quantification, albumin quantification, immunohistochemistry, and RT-qPCR gene expression analysis. Results showed significant increases in proliferative activity of HepG2 on rLECM containing scaffolds alongside maintained key gene expression. This study confirms that rLECM can be utilized to modulate the bioactivity of electrospun PCL scaffolds and has the potential to produce electrospun scaffolds suitable for enhanced hepatocyte cultures and in-vitro liver tissue models.
Collapse
Affiliation(s)
- Thomas S. R. Bate
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| | - William Shanahan
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| | - Joseph P. Casillo
- School of GeoSciencesUniversity of Edinburgh, Grant InstituteEdinburghUK
| | - Rhiannon Grant
- MERLN InstituteMaastricht UniversityMaastrichtThe Netherlands
| | - Stuart J. Forbes
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | - Anthony Callanan
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| |
Collapse
|
8
|
Bianchi S, Bernardi S, Simeone D, Torge D, Macchiarelli G, Marchetti E. Proliferation and Morphological Assessment of Human Periodontal Ligament Fibroblast towards Bovine Pericardium Membranes: An In Vitro Study. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15238284. [PMID: 36499781 PMCID: PMC9740786 DOI: 10.3390/ma15238284] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 05/13/2023]
Abstract
Over the past decade regenerative branches of dentistry have taken on more and more importance, resulting in the development of performing scaffold materials. These should induce cell adhesion, support, and guide the tissues’ growth. Among the developed materials, we can include resorbable or non-membranes. The purpose of this study was to investigate the proliferation abilities and the attachment of human periodontal ligament fibroblasts (HPLIFs) over two bovine pericardium membranes with different thicknesses, 0.2 mm and 0.4 mm, respectively. These membranes have been decellularized by the manufacturer, preserving the three-dimensional collagen’s structure. The HPLFs were cultured in standard conditions and exposed to the tested materials. XTT was performed to assess cell proliferation, while light microscopy (LM) and scanning electron microscopy (SEM) observations assessed fibroblast morphology at different times (T1, T2, and T3). Proliferation assays have shown a statistically significant difference in growth at T1 (p < 0.05) in the cells cultured with a thicker membrane compared to the thinner one. LM analysis showed healthy fibroblasts in contact with the membranes, appearing larger and with a polygonal shape. SEM observation demonstrated thickening of the fibroblasts which continued to adhere to the membrane’s surface, with enlarged polygonal shape and developed filipodia and lamellipodia. These results showed a similar cell behavior over the two bovine pericardium membranes, demonstrating a cellular migration along and within the layers of the membrane, binding with membrane fibers by means of filopodial extensions. Knowledge of the effects of the collagen membranes derived from bovine pericardium on cellular behavior will help clinicians choose the type of scaffolds according to the required clinical situation.
Collapse
|
9
|
Cavanaugh M, Asheghali D, Motta CM, Silantyeva E, Nikam SP, Becker ML, Willits RK. Influence of Touch-Spun Nanofiber Diameter on Contact Guidance during Peripheral Nerve Repair. Biomacromolecules 2022; 23:2635-2646. [DOI: 10.1021/acs.biomac.2c00379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- McKay Cavanaugh
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Darya Asheghali
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Cecilia M. Motta
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Elena Silantyeva
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Shantanu P. Nikam
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Matthew L. Becker
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
- Department of Orthopaedic Surgery, Duke University, Durham, North Carolina 27708, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Rebecca K. Willits
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
10
|
Xia J, Liu ZY, Han ZY, Yuan Y, Shao Y, Feng XQ, Weitz DA. Regulation of cell attachment, spreading, and migration by hydrogel substrates with independently tunable mesh size. Acta Biomater 2022; 141:178-189. [PMID: 35041902 PMCID: PMC8898306 DOI: 10.1016/j.actbio.2022.01.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/25/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Hydrogels are widely used as substrates to investigate interactions between cells and their microenvironment as they mimic many attributes of the extracellular matrix. The stiffness of hydrogels is an important property that is known to regulate cell behavior. Beside stiffness, cells also respond to structural cues such as mesh size. However, since the mesh size of hydrogel is intrinsically coupled to its stiffness, its role in regulating cell behavior has never been independently investigated. Here, we report a hydrogel system whose mesh size and stiffness can be independently controlled. Cell behavior, including spreading, migration, and formation of focal adhesions is significantly altered on hydrogels with different mesh sizes but with the same stiffness. At the transcriptional level, hydrogel mesh size affects cellular mechanotransduction by regulating nuclear translocation of yes-associated protein. These findings demonstrate that the mesh size of a hydrogel plays an important role in cell-substrate interactions. STATEMENT OF SIGNIFICANCE: Hydrogels are ideal platforms with which to investigate interactions between cells and their microenvironment as they mimic many physical properties of the extracellular matrix. However, the mesh size of hydrogels is intrinsically coupled to their stiffness, making it challenging to investigate the contribution of mesh size to cell behavior. In this work, we use hydrogel-on-glass substrates with defined thicknesses whose stiffness and mesh size can be independently tuned. We use these substrates to isolate the effects of mesh size on cell behavior, including attachment, spreading, migration, focal adhesion formation and YAP localization in the nucleus. Our results show that mesh size has significant, yet often overlooked, effects, on cell behavior, and contribute to a further understanding of cell-substrate interactions.
Collapse
Affiliation(s)
- Jing Xia
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Zong-Yuan Liu
- Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing 100084, China
| | - Zheng-Yuan Han
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Yuan Yuan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Yue Shao
- Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing 100084, China
| | - Xi-Qiao Feng
- Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing 100084, China.
| | - David A Weitz
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
11
|
Three-dimensional models: a novel approach for lymphoma research. J Cancer Res Clin Oncol 2022; 148:753-765. [DOI: 10.1007/s00432-021-03897-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022]
|
12
|
Zhang Y, Zhang M, Cheng D, Xu S, Du C, Xie L, Zhao W. Applications of electrospun scaffolds with enlarged pores in tissue engineering. Biomater Sci 2022; 10:1423-1447. [DOI: 10.1039/d1bm01651b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite electrospinning has multiple advantages over other methods such as creating materials with superfine fiber diameter, high specific surface area, and good mechanical properties, the pore diameter of scaffolds prepared...
Collapse
|
13
|
Bosworth LA, Lanaro M, O'Loughlin DA, D'Sa RA, Woodruff MA, Williams RL. Melt electro-written scaffolds with box-architecture support orthogonally oriented collagen. Biofabrication 2021; 14. [PMID: 34883476 DOI: 10.1088/1758-5090/ac41a1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 12/09/2021] [Indexed: 11/12/2022]
Abstract
Melt electro-writing (MEW) is a state-of-the-art technique that supports fabrication of 3D, precisely controlled and reproducible fiber structures. A standard MEW scaffold design is a box-structure, where a repeat layer of 90° boxes is produced from a single fiber. In 3D form (i.e. multiple layers), this structure has the potential to mimic orthogonal arrangements of collagen, as observed in the corneal stroma. In this study, we determined the response of human primary corneal stromal cells and their deposited fibrillar collagen (detected using a CNA35 probe) following six weeksin vitroculture on these box-structures made from poly(ϵ-caprolactone) (PCL). Comparison was also made to glass substrates (topography-free) and electrospun PCL fibers (aligned topography). Cell orientation and collagen deposition were non-uniform on glass substrates. Electrospun scaffolds supported an excellent parallel arrangement of cells and deposited collagen to the underlying architecture of aligned fibers, but there was no evidence of bidirectional collagen. In contrast, MEW scaffolds encouraged the formation of a dense, interconnected cellular network and deposited fibrillar collagen layers with a distinct orthogonal-arrangement. Collagen fibrils were particularly dominant through the middle layers of the MEW scaffolds' total thickness and closer examination revealed these fibrils to be concentrated within the pores' central regions. With the demand for donor corneas far exceeding the supply-leaving many with visual impairment-the application of MEW as a potential technique to recreate the corneal stroma with spontaneous, bidirectional collagen organization warrants further study.
Collapse
Affiliation(s)
- Lucy A Bosworth
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Matthew Lanaro
- Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Danielle A O'Loughlin
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Raechelle A D'Sa
- Department of Mechanical, Materials and Aerospace Engineering, Faculty of Science and Engineering, University of Liverpool, Liverpool L69 3GH, United Kingdom
| | - Maria A Woodruff
- Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Rachel L Williams
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| |
Collapse
|
14
|
Mohseni Garakani M, Ahangar P, Watson S, Nisol B, Wertheimer MR, Rosenzweig DH, Ajji A. A novel 3D co-culture platform for integrating tissue interfaces for tumor growth, migration and therapeutic sensitivity: “PP-3D-S”. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112566. [DOI: 10.1016/j.msec.2021.112566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023]
|
15
|
The Effect of Strontium-Substituted Hydroxyapatite Nanofibrous Matrix on Osteoblast Proliferation and Differentiation. MEMBRANES 2021; 11:membranes11080624. [PMID: 34436387 PMCID: PMC8401295 DOI: 10.3390/membranes11080624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/20/2022]
Abstract
Natural bone tissue consists primarily of bioapatite and collagen. Synthetic hydroxyapatite (HA) possesses good biocompatibility, bioactivity, and osteoconductivity due to its chemical and biological similarity to bioapatite. Hence, HA has been widely used as a bone graft, cell carrier and drug/gene delivery carrier. Moreover, strontium-substituted hydroxyapatite (SrHA) can enhance osteogenic differentiation and inhibit adipogenic differentiation of mesenchymal stem cells. Hence, SrHA has the potential to be used as a bone graft for bone regeneration. It is widely accepted that cell adhesion and most cellular activities are sensitive to the topography and molecular composition of the matrix. Electrospun polymer or polymer-bioceramic composite nanofibers have been demonstrated to enhance osteoblast differentiation. However, to date, no studies have investigated the effect of nanofibrous bioceramic matrices on osteoblasts. In this study, hydroxyapatite nanofiber (HANF) and strontium-substituted hydroxyapatite nanofiber (SrHANF) matrices were fabricated by electrospinning. The effect of the HANF components on MG63 osteoblast-like cells was evaluated by cell morphology, proliferation, alkaline phosphatase activity (ALP) and gene expression levels of RUNX2, COLI, OCN and BSP. The results showed that MG63 osteoblast-like cells exhibited higher ALP and gene expression levels of RUNX2, COLI, BSP and OCN on the SrHANF matrix than the HANF matrix. Hence, SrHANFs could enhance the differentiation of MG63 osteoblast-like cells.
Collapse
|
16
|
Miri V, Asadi A, Sagha M, Najafzadeh N, Golmohammadi MG. Poly (L-lactic acid) nanofibrous scaffolds support the proliferation and neural differentiation of mouse neural stem and progenitor cells. Int J Dev Neurosci 2021; 81:438-447. [PMID: 33934403 DOI: 10.1002/jdn.10119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The distribution and growth of cells on nanofibrous scaffolds seem to be an indispensable precondition in cell tissue engineering. The potential use of biomaterial scaffolds in neural stem cell therapy is increasingly attracting attention. AIM In this study, we produced porous nanofibrous scaffolds fabricated from random poly-L-lactic acid (PLLA) to support neurogenic differentiation of neural stem and progenitor cells (NSPCs), isolated from the subventricular zone (SVZ) of the adult mouse brain. METHODS The viability and proliferation of the NSPCs on the nanofibrous PLLA scaffold were also tested by nuclear staining with 4, 6-diamidino-2-phenylindole dihydrochloride (DAPI), 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay and scanning electron microscopy (SEM). To investigate the differentiation potential of NSPCs on the scaffolds, the cells were treated with a neurogenic differentiation medium, and immunostaining was done to detect neuronal and glial cells after 14 and 21 days of cultivation. Furthermore, the morphology of differentiated cells on the scaffold was examined using SEM. RESULTS The DAPI staining revealed the proliferation of NSPCs onto the surface of the nanofibrous PLLA scaffold. DAPI-positive cells were counted on days 2 and 5 after cultivation. The mean number of cells in each microscopic field was significantly (p < .05) increased (51 ± 19 on day 2 compared to 77 ± 25 cells on day 5). The results showed that the cell viability on PLLA scaffolds significantly increased compared to control groups. Moreover, cell viability was significantly increased 5 days after culturing (262.3 ± 50.2) as compared to 2 days culture in Vitro (174.2 ± 28.3, p < .05). Scanning electron micrographs also showed that the NSPCs adhered and differentiated on PLLA scaffolds. We found that the neural cell markers, microtubule-associated protein 2 (MAP2) and glial fibrillary acidic protein (GFAP), were expressed in NSPCs seeded on random PLLA scaffolds after 21 days of cultivation. CONCLUSION These results suggest that the PLLA nano-scaffolds, due to their biocompatible property, are an appropriate structure for the proliferation, differentiation, and normal growth of NSPCs.
Collapse
Affiliation(s)
- Vahideh Miri
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Asadollah Asadi
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mohsen Sagha
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nowruz Najafzadeh
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Ghasem Golmohammadi
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
17
|
Ghanbari E, Solouk A, Mehdinavaz Aghdam R, Haghbin Nazarpak M, Ahmadi Tafti SH. A novel substrate based on electrospun polyurethane nanofibers and electrosprayed polyvinyl alcohol microparticles for recombinant human erythropoietin delivery. J Biomed Mater Res A 2021; 110:181-195. [PMID: 34309172 DOI: 10.1002/jbm.a.37275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/29/2021] [Accepted: 07/07/2021] [Indexed: 11/08/2022]
Abstract
After myocardial infarction caused by a heart attack, endothelial cells need to be preserved in order to regenerate new capillaries. Moreover, sufficient mechanical support is necessary for the infarcted myocardium to pump the blood. Herein, we designed a novel substrate containing polyurethane (PU) nanofibrous layers and recombinant human erythropoietin (rhEPO)-loaded microparticles for both controlled releases of rhEPO and mechanical support of myocardium. In this system, the single-layer (SL) and double-layer (DL) PU nanofibers were electrospun, and then microparticles with different rhEPO:polyvinyl alcohol (PVA) ratios were electrosprayed on the layers. The in vitro release behavior of rhEPO from SL substrates was not satisfactory, and then the study focused on DL patches in which the release profile was in accordance with Korsmeyer-Peppas model. The release exponent of 0.89 for the DL PU/120PVA:1rhEPO represented zero-order release. The results inferred that these substrates possessed highly tailored mechanical properties; Young's modulus and ultimate tensile strength of the substrates were 74-172 kPa and 7.4-9.9 MPa, respectively. The rhEPO release from the substrates was leading to the proper adhesion of endothelial cells and more than 95% cell viability. The results indicated that the patch of elastic nanofibers and microparticles offered a potential substrate for simultaneous rhEPO delivery to endothelial cells and also mechanically supporting the infarcted myocardium.
Collapse
Affiliation(s)
- Elmira Ghanbari
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Atefeh Solouk
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | | | - Masoumeh Haghbin Nazarpak
- New Technologies Research Center, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | | |
Collapse
|
18
|
Adhikari KR, Stanishevskaya I, Caracciolo PC, Abraham GA, Thomas V. Novel Poly(ester urethane urea)/Polydioxanone Blends: Electrospun Fibrous Meshes and Films. Molecules 2021; 26:3847. [PMID: 34202602 PMCID: PMC8270292 DOI: 10.3390/molecules26133847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 11/22/2022] Open
Abstract
In this work, we report the electrospinning and mechano-morphological characterizations of scaffolds based on blends of a novel poly(ester urethane urea) (PHH) and poly(dioxanone) (PDO). At the optimized electrospinning conditions, PHH, PDO and blend PHH/PDO in Hexafluroisopropanol (HFIP) solution yielded bead-free non-woven random nanofibers with high porosity and diameter in the range of hundreds of nanometers. The structural, morphological, and biomechanical properties were investigated using Differential Scanning Calorimetry, Scanning Electron Microscopy, Atomic Force Microscopy, and tensile tests. The blended scaffold showed an elastic modulus (~5 MPa) with a combination of the ultimate tensile strength (2 ± 0.5 MPa), and maximum elongation (150% ± 44%) in hydrated conditions, which are comparable to the materials currently being used for soft tissue applications such as skin, native arteries, and cardiac muscles applications. This demonstrates the feasibility of an electrospun PHH/PDO blend for cardiac patches or vascular graft applications that mimic the nanoscale structure and mechanical properties of native tissue.
Collapse
Affiliation(s)
- Kiran R. Adhikari
- Department of Physics, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Center for Nanoscale Materials and Biointegration (CNMB), University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Pablo C. Caracciolo
- Instituto de Investigaciones en Ciencia y Tecnología de Materiales, INTEMA (UNMdP-CONICET), Av. Juan B. Justo 4302, B7608FDQ Mar del Plata, Argentina; (P.C.C.); (G.A.A.)
| | - Gustavo A. Abraham
- Instituto de Investigaciones en Ciencia y Tecnología de Materiales, INTEMA (UNMdP-CONICET), Av. Juan B. Justo 4302, B7608FDQ Mar del Plata, Argentina; (P.C.C.); (G.A.A.)
| | - Vinoy Thomas
- Center for Nanoscale Materials and Biointegration (CNMB), University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Materials Science and Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
19
|
Fadil F, Affandi NDN, Misnon MI, Bonnia NN, Harun AM, Alam MK. Review on Electrospun Nanofiber-Applied Products. Polymers (Basel) 2021; 13:2087. [PMID: 34202857 PMCID: PMC8271930 DOI: 10.3390/polym13132087] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 11/17/2022] Open
Abstract
Electrospinning technology, which was previously known as a scientific interdisciplinary research approach, is now ready to move towards a practice-based interdisciplinary approach in a variety of fields, progressively. Electrospun nanofiber-applied products are made directly from a nonwoven fabric-based membranes prepared from polymeric liquids involving the application of sufficiently high voltages during electrospinning. Today, electrospun nanofiber-based materials are of remarkable interest across multiple fields of applications, such as in electronics, sensors, functional garments, sound proofing, filters, wound dressing and scaffolds. This article presents such a review for summarizing the current progress on the manufacturing scalability of electrospun nanofibers and the commercialization of electrospun nanofiber products by dedicated companies globally. Despite the clear potential and limitless possibilities for electrospun nanofiber applications, the uptake of electrospinning by the industry is still limited due to the challenges in the manufacturing and turning of electrospun nanofibers into physical products. The recent developments in the field of electrospinning, such as the prominent nonwoven technology, personal views and the potential path forward for the growth of commercially applied products based on electrospun nanofibers, are also highlighted.
Collapse
Affiliation(s)
- Fatirah Fadil
- Textile Research Group, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Selangor, Malaysia; (F.F.); (M.I.M.)
| | - Nor Dalila Nor Affandi
- Textile Research Group, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Selangor, Malaysia; (F.F.); (M.I.M.)
| | - Mohd Iqbal Misnon
- Textile Research Group, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Selangor, Malaysia; (F.F.); (M.I.M.)
| | - Noor Najmi Bonnia
- Materials Science and Technology, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Selangor, Malaysia;
| | - Ahmad Mukifza Harun
- Faculty Engineering, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | | |
Collapse
|
20
|
Conversion of Electrospun Chitosan into Chitin: A Robust Strategy to Tune the Properties of 2D Biomimetic Nanofiber Scaffolds. POLYSACCHARIDES 2021. [DOI: 10.3390/polysaccharides2020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
New biomimetic micro- and nano-CsU-based fibrous scaffolds electrospun from solution containing high purity-medical grade chitosan (CsU) of fungus origin (CsU1, Mv ~174,000 and CsU2, 205,000, degree of deacetylation (DDA) ~65%) and polyethylene oxide (PEO, Mv ~ 900,000), in the presence of given amounts of Triton X-100 (from 0.01 to 0.5 wt%) as surfactant were fabricated. We demonstrate that by carefully selecting compositions and surfactant levels, porous mats with CsU content up to 90% (at this molecular weight and DDA) were achieved. Remarkable long-term stability in water or phosphate buffer solution storage were obtained by developing post-electrospinning treatment allowing the complete elimination of the PEO from the CsU-fibers as demonstrated by TGA, DSC and ESEM analysis. Subsequent reacetylation procedure was applied to convert 2D biomimetic chitosan mats to chitin (CsE)-based ones while preserving the nanofiber structure. This innovative procedure allows tuning and modifying the thermal, mechanical properties and more importantly the biodegradation abilities (fast enzymatic biodegradation in some cases and slower on the others) of the prepared nanofibrous mats. The established reproducible method offers the unique advantage to modulate the membrane properties leading to stable 2D biomimetic CsU and/or chitin (CsE) scaffolds tailor-made for specific purposes in the field of tissue engineering.
Collapse
|
21
|
Lee J, Lee S, Kim SM, Shin H. Size-controlled human adipose-derived stem cell spheroids hybridized with single-segmented nanofibers and their effect on viability and stem cell differentiation. Biomater Res 2021; 25:14. [PMID: 33902733 PMCID: PMC8074457 DOI: 10.1186/s40824-021-00215-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Fabrication of three-dimensional stem cell spheroids have been studied to improve stem cell function, but the hypoxic core and limited penetration of nutrients and signaling cues to the interior of the spheroid were challenges. The incorporation of polymers such as silica and gelatin in spheroids resulted in relatively relaxed assembly of composite spheroids, and enhancing transport of nutrient and biological gas. However, because of the low surface area between cells and since the polymers were heterogeneously distributed throughout the spheroid, these polymers cannot increase the cell to extracellular matrix interactions needed to support differentiation. METHODS We developed the stem cell spheroids that incorporate poly(ι-lactic acid) single-segmented fibers synthesized by electrospinning and physical and chemical fragmentation. The proper mixing ratio was 2000 cells/μg fibers (average length of the fibers was 50 μm - 100 μm). The SFs were coated with polydopamine to increase cell binding affinity and to synthesize various-sized spheroids. The function of spheroids was investigated by in vitro analysis depending on their sizes. For statistical analysis, Graphpad Prism 5 software (San Diego, CA, USA) was used to perform one-way analysis of variance ANOVA with Tukey's honest significant difference test and a Student's t-test (for two variables) (P < 0.05). RESULTS Spheroids of different sizes were created by modulating the amount of cells and fibers (0.063 mm2-0.322 mm2). The fibers in the spheroid were homogenously distributed and increased cell viability, while cell-only spheroids showed a loss of DNA contents, internal degradation, and many apoptotic signals. Furthermore, we investigated stemness and various functions of various-sized fiber-incorporated spheroids. In conclusion, the spheroid with the largest size showed the greatest release of angiogenic factors (released VEGF: 0.111 ± 0.004 pg/ng DNA), while the smallest size showed greater effects of osteogenic differentiation (mineralized calcium: 18.099 ± 0.271 ng/ng DNA). CONCLUSION The spheroids incorporating polydopamine coated single-segmented fibers showed enhanced viability regardless of sizes and increased their functionality by regulating the size of spheroids which may be used for various tissue reconstruction and therapeutic applications.
Collapse
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- BK21 FOUR, Human-Tech Convergence Program, Hanyang University, Seoul, 04763, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, 04763, Republic of Korea
| | - Sung Min Kim
- BK21 FOUR, Human-Tech Convergence Program, Hanyang University, Seoul, 04763, Republic of Korea.
- Department of Physical Education and Active Aging Industry, Hanyang University, Seoul, 04763, Republic of Korea.
- Center for Artificial Intelligence Muscle, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, 04763, Republic of Korea.
- Institute of Nano Science and Technology, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
22
|
Hong J, Shin Y, Lee J, Cha C. Programmable multilayer printing of a mechanically-tunable 3D hydrogel co-culture system for high-throughput investigation of complex cellular behavior. LAB ON A CHIP 2021; 21:710-718. [PMID: 33459335 DOI: 10.1039/d0lc01230k] [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
Hydrogels are widely used as a 3D cell culture platform, as they can be tailored to provide suitable microenvironments to induce cellular phenotypes with physiological significance. Hydrogels are especially deemed attractive as a co-culture platform, in which two or more different types of cells are cultured together in close proximity, since the spatial distribution of different cell types can be rendered possible by advanced microfabrication schemes. Herein, programmable multilayer photolithography is employed to develop a 3D hydrogel-based co-culture system in an efficient and scalable manner, which consists of an inner microgel array containing one cell type covered by an outer hydrogel overlay containing another cell type. In particular, the mechanical properties of microgel array and hydrogel overlay are independently controlled in a wide range, with elastic moduli ranging from 1.7 to 31.6 kPa, allowing the high-throughput investigation of both individual hydrogel mechanics and mechanical gradients generated at their interface. Utilizing this system, phenotypical changes (i.e. proliferation, spheroid formation and Mφ polarization) of macrophages encapsulated in microgel array, in response to complex mechanical microenvironment and co-cultured fibroblasts, are comprehensively explored.
Collapse
Affiliation(s)
- Jisu Hong
- Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea. and Center for Multidimensional Programmable Matter, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea
| | - Yoonkyung Shin
- Department of Energy Engineering, School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea.
| | - Jiseok Lee
- Center for Multidimensional Programmable Matter, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea and Department of Energy Engineering, School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea.
| | - Chaenyung Cha
- Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea. and Center for Multidimensional Programmable Matter, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea
| |
Collapse
|
23
|
Saydé T, El Hamoui O, Alies B, Gaudin K, Lespes G, Battu S. Biomaterials for Three-Dimensional Cell Culture: From Applications in Oncology to Nanotechnology. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:481. [PMID: 33668665 PMCID: PMC7917665 DOI: 10.3390/nano11020481] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Three-dimensional cell culture has revolutionized cellular biology research and opened the door to novel discoveries in terms of cellular behavior and response to microenvironment stimuli. Different types of 3D culture exist today, including hydrogel scaffold-based models, which possess a complex structure mimicking the extracellular matrix. These hydrogels can be made of polymers (natural or synthetic) or low-molecular weight gelators that, via the supramolecular assembly of molecules, allow the production of a reproducible hydrogel with tunable mechanical properties. When cancer cells are grown in this type of hydrogel, they develop into multicellular tumor spheroids (MCTS). Three-dimensional (3D) cancer culture combined with a complex microenvironment that consists of a platform to study tumor development and also to assess the toxicity of physico-chemical entities such as ions, molecules or particles. With the emergence of nanoparticles of different origins and natures, implementing a reproducible in vitro model that consists of a bio-indicator for nano-toxicity assays is inevitable. However, the maneuver process of such a bio-indicator requires the implementation of a repeatable system that undergoes an exhaustive follow-up. Hence, the biggest challenge in this matter is the reproducibility of the MCTS and the associated full-scale characterization of this system's components.
Collapse
Affiliation(s)
- Tarek Saydé
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, 87025 Limoges, France;
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
| | - Omar El Hamoui
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
- CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux (IPREM), UMR 5254, Université de Pau et des Pays de l’Adour (E2S/UPPA), 2 Avenue Pierre Angot, 64053 Pau, France
| | - Bruno Alies
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
| | - Karen Gaudin
- ARNA, INSERM U1212, UMR CNRS 5320, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; (O.E.H.); (B.A.); (K.G.)
| | - Gaëtane Lespes
- CNRS, Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux (IPREM), UMR 5254, Université de Pau et des Pays de l’Adour (E2S/UPPA), 2 Avenue Pierre Angot, 64053 Pau, France
| | - Serge Battu
- EA3842-CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, 87025 Limoges, France;
| |
Collapse
|
24
|
Kwon T, Sung BJ. Confinement effects on the mechanical heterogeneity of polymer fiber glasses. Phys Rev E 2020; 102:052501. [PMID: 33327119 DOI: 10.1103/physreve.102.052501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/13/2020] [Indexed: 11/07/2022]
Abstract
Both polymer fiber glasses and bulk polymer glasses exhibit nonlinear mechanical responses under uniaxial deformation. In polymer fibers, however, polymer chains are confined strongly and the surface area is relatively large compared to their volume. The confinement and the surface may lead to the spatially heterogeneous relaxation of chains in polymer fibers. In this work we perform molecular dynamics simulations and investigate the relation between the heterogeneous dynamics and the nonlinear mechanical responses at a molecular level. Our molecular simulations capture successfully not only the nonlinear mechanical response but also the dependence of mechanical properties on the strain rate of typical polymer glasses as in experiments. We find that the local elastic modulus and the nonaffine displacement are spatially heterogeneous in the pre-yield regime, which results in a lower elastic modulus for polymer fibers than bulk polymer glasses. In the post-yield regime, those mechanical properties become relatively homogeneous. Monomers with large nonaffine displacement are localized mainly at the interfacial region in the pre-yield regime while highly nonaffine monomers are distributed throughout the fibers in the post-yield regime. We show that the nonaffine displacement during deformation relates closely to the mechanical response of the polymer fibers. We also find that in the strain-hardening regime there is a significant difference in the energetic contribution to the stress between polymer fibers and bulk polymers, for which the modulus of the strain-hardening regime of the polymer fibers is smaller than that of bulk polymers.
Collapse
Affiliation(s)
- Taejin Kwon
- Department of Chemistry, Sogang University, Seoul 04107, Republic of Korea
| | - Bong June Sung
- Department of Chemistry, Sogang University, Seoul 04107, Republic of Korea
| |
Collapse
|
25
|
Tang H, Yi B, Wang X, Shen Y, Zhang Y. Understanding the cellular responses based on low-density electrospun fiber networks. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111470. [PMID: 33321594 DOI: 10.1016/j.msec.2020.111470] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/14/2020] [Accepted: 08/25/2020] [Indexed: 01/12/2023]
Abstract
Fibers produced from electrospinning are well-known to be extremely fine with diameters ranging from tens of nanometers to a few microns. Such ultrafine fibers not only allow for engineering scaffolds resembling the ultrastructure of the native extracellular matrix, but also offer possibility to explore the remodeling behavior of cells in vitro, due to their mechanically 'adequate' softness endowed by their ultrafine fineness. However, the remodeling effect of cells on the biomimicking fibrous substrates remains to be understood, because the crisscrossing and entangling among nanofibers in those tightly packed fibrous mats ultimately lead to merely a topological phenomenon, similar to that of the nanofiber-like topography embossed on the surface of a solid matter. In this study, the effect of nanofiber density on cellular response behavior was investigated by reducing the density of electrospun fiber networks. Using polycaprolactone (PCL) as a model polymer, randomly oriented fiber networks with various densities, namely, 37.7 ± 16.3 μg/cm2 (D1), 103.8 ± 16.3 μg/cm2 (D2), 198.2 ± 40.0 μg/cm2 (D3), and 471.8 ± 32.7 μg/cm2 (D4), were prepared by electrospinning for varied collection durations (10 s, 50 s, 100 s, and 10 min, respectively). By examining the responsive behavior of the human induced pluripotent stem cell-derived mesenchymal stem cells (hiPS-MSCs) cultured on these nanofibrous networks, we showed that the fiber network with a moderate density (D2) is beneficial to the cell attachment, spreading, actin polymerization, contractility and migration. There also showed an increased tendency in nuclear localization of the Yes-associated protein (YAP) and subsequent activation of YAP responsive gene transcription, and cell proliferation and collagen synthesis were also enhanced on the D2. However, further increasing the fiber density (D3, D4) gave rise to weakened induction effect of fibers on the cellular responses. These results enrich our understanding on the effect of fiber density on cell behavior, and disclose the dependence of cellular responses on fiber density. This study paves the way to precisely design biomimetic fibrous scaffolds for achieving enhanced cell-scaffold interactions and tissue regeneration.
Collapse
Affiliation(s)
- Han Tang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Bingcheng Yi
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Xianliu Wang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Yanbing Shen
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Yanzhong Zhang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China; Key Lab of Science & Technology of Eco-Textile, Ministry of Education, Donghua University, Shanghai 201620, China; Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China.
| |
Collapse
|
26
|
Chandika P, Heo SY, Kim TH, Oh GW, Kim GH, Kim MS, Jung WK. Recent advances in biological macromolecule based tissue-engineered composite scaffolds for cardiac tissue regeneration applications. Int J Biol Macromol 2020; 164:2329-2357. [DOI: 10.1016/j.ijbiomac.2020.08.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
|
27
|
Abstract
Polylactic acid (PLA) nanofiber nonwovens have recently come under more vigorous investigation for their use as tissue engineering scaffolds owing to its ability to mimic the physical properties of naturally occurring human extracellular matrix in a variety of host tissues. Currently, the majority of available research on PLA nanowebs has focused on their creation through electrospinning. The goal of this study was to evaluate meltblown nonwoven webs made of nanodiameter PLA fibers for their application as a tissue engineering scaffold. Meltblown PLA fabrics were produced with a variety of different crystallinities, tensile moduli, and pore diameters. One fabric with mechanical properties similar to human dermis was selected as a scaffold to study attachment, proliferation, and migration of human dermal fibroblasts over 1, 3, 7, and 14 days without the use of additional cell adhesion molecules. The 3-(4,5-dimethylthiazol-2-yl)-diphenyltetrazolium bromide assay showed good proliferation from day 1 to 3 (P = 0.026) and up to 7 days of culture (P = 0.005) but without increase from day 7 to 14. Electron microscopy demonstrated adequate cellular attachment and surface migration at 1, 3, 7, and 14 days. Finally, confocal microscopy was used to investigate cellular penetration into the scaffolds. The investigation found that cells were able to penetrate fully through the thickness of the scaffold. The successes of this initial experiment are promising and confirm that meltblown nanofiber nonwovens are a viable avenue for tissue engineering scaffolds. Hopefully, these conclusions will open the door for others to pursue research in this exciting field.
Collapse
|
28
|
Guo B, Tang C, Wang M, Zhao Z, Shokoohi‐Tabrizi HA, Shi B, Andrukhov O, Rausch‐Fan X. In vitro biocompatibility of biohybrid polymers membrane evaluated in human gingival fibroblasts. J Biomed Mater Res B Appl Biomater 2020; 108:2590-2598. [PMID: 32096606 PMCID: PMC7383566 DOI: 10.1002/jbm.b.34591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/11/2020] [Indexed: 12/14/2022]
Abstract
The biohybrid polymer membrane (BHM) is a new biomaterial designed for the treatment of soft periodontal tissue defects. We aimed to evaluate the in vitro biocompatibility of the membrane in human gingival fibroblasts and the capability to induce cell adhesion, migration, differentiation and improving the production of the extracellular matrix. BHM and Mucograft® collagen matrix (MCM) membranes were punched into 6 mm diameter round discs and placed in 96-well plates. Human primary gingival fibroblasts were seeded on the membranes or tissue culture plastic (TCP) serving as the control. Cell proliferation/viability and morphology were evaluated after 3, 7, and 14 days of culture by cell counting kit (CCK)-8 assay and scanning electron microscopy, respectively. Additionally, the gene expression of transforming growth factor (TGF)-β1, focal adhesion kinase (FAK), collagen type 1 (Col1), alpha-smooth muscle actin (α-SMA), and fibroblasts growth factor (FGF)-2 was analyzed at 3, 7, and 14 days of culture by qPCR. Cell proliferation on BHM was significantly higher than on MCM and similar to TCP. Gene expression of TGF-β1, FAK, Col1, and α-SMA were significantly increased on BHM compared to TCP at most investigated time points. However, the gene expression of FGF-2 was significantly decreased on BHM at Day 7 and recovered at Day 14 to the levels similar to TCP. The finding of this study showed that BHM is superior for gingival fibroblasts in terms of adhesion, proliferation, and gene expression, suggesting that this membrane may promote the healing of soft periodontal tissue.
Collapse
Affiliation(s)
- Bin Guo
- Department of StomatologyJinan Central Hospital affiliated to Shandong UniversityJinanShandongChina
- Division of Periodontology and Conservative DentistryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Chuhua Tang
- Division of Periodontology and Conservative DentistryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Department of StomatologyPLA Strategic Support Force Characteristic Medical CenterBeijingChina
| | - Mingguo Wang
- Department of StomatologyJinan Central Hospital affiliated to Shandong UniversityJinanShandongChina
| | - Zhongqi Zhao
- Division of Periodontology and Conservative DentistryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Hassan A. Shokoohi‐Tabrizi
- Division of Periodontology and Conservative DentistryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Bin Shi
- Department of Oral and Maxillofacial SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| | - Oleh Andrukhov
- Division of Periodontology and Conservative DentistryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Xiaohui Rausch‐Fan
- Division of Periodontology and Conservative DentistryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Department of Oral and Maxillofacial SurgeryThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
29
|
Potential Implantable Nanofibrous Biomaterials Combined with Stem Cells for Subchondral Bone Regeneration. MATERIALS 2020; 13:ma13143087. [PMID: 32664278 PMCID: PMC7412392 DOI: 10.3390/ma13143087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/28/2022]
Abstract
The treatment of osteochondral defects remains a challenge. Four scaffolds were produced using Food and Drug Administration (FDA)-approved polymers to investigate their therapeutic potential for the regeneration of the osteochondral unit. Polycaprolactone (PCL) and poly(vinyl-pyrrolidone) (PVP) scaffolds were made by electrohydrodynamic techniques. Hydroxyapatite (HAp) and/or sodium hyaluronate (HA) can be then loaded to PCL nanofibers and/or PVP particles. The purpose of adding hydroxyapatite and sodium hyaluronate into PCL/PVP scaffolds is to increase the regenerative ability for subchondral bone and joint cartilage, respectively. Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) were seeded on these biomaterials. The biocompatibility of these biomaterials in vitro and in vivo, as well as their potential to support MSC differentiation under specific chondrogenic or osteogenic conditions, were evaluated. We show here that hBM-MSCs could proliferate and differentiate both in vitro and in vivo on these biomaterials. In addition, the PCL-HAp could effectively increase the mineralization and induce the differentiation of MSCs into osteoblasts in an osteogenic condition. These results indicate that PCL-HAp biomaterials combined with MSCs could be a beneficial candidate for subchondral bone regeneration.
Collapse
|
30
|
Rao GSNK, Kurakula M, Yadav KS. Application of Electrospun Materials in Gene Delivery. ELECTROSPUN MATERIALS AND THEIR ALLIED APPLICATIONS 2020:265-306. [DOI: 10.1002/9781119655039.ch10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
31
|
Gritsch L, Liverani L, Lovell C, Boccaccini AR. Polycaprolactone Electrospun Fiber Mats Prepared Using Benign Solvents: Blending with Copper(II)‐Chitosan Increases the Secretion of Vascular Endothelial Growth Factor in a Bone Marrow Stromal Cell Line. Macromol Biosci 2020; 20:e1900355. [DOI: 10.1002/mabi.201900355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/08/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Lukas Gritsch
- Institute of BiomaterialsUniversity of Erlangen‐Nuremberg Cauerstraße 6 91058 Erlangen Germany
- Lucideon Ltd. Queens Road, Penkhull Stoke‐on‐Trent Staffordshire ST4 7LQ UK
| | - Liliana Liverani
- Institute of BiomaterialsUniversity of Erlangen‐Nuremberg Cauerstraße 6 91058 Erlangen Germany
| | - Christopher Lovell
- Lucideon Ltd. Queens Road, Penkhull Stoke‐on‐Trent Staffordshire ST4 7LQ UK
| | - Aldo R. Boccaccini
- Institute of BiomaterialsUniversity of Erlangen‐Nuremberg Cauerstraße 6 91058 Erlangen Germany
| |
Collapse
|
32
|
Tahmasebi A, Enderami SE, Saburi E, Islami M, Yaslianifard S, Mahabadi JA, Ardeshirylajimi A, Soleimanifar F, Moghadam AS. Micro-RNA-incorporated electrospun nanofibers improve osteogenic differentiation of human-induced pluripotent stem cells. J Biomed Mater Res A 2020; 108:377-386. [PMID: 31654461 DOI: 10.1002/jbm.a.36824] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/20/2022]
Abstract
Smart scaffolds have a great role in the damaged tissue reconstruction. The aim of this study was developing a scaffold that in addition to its fiber's topography has also content of micro-RNAs (miRNAs), which play a regulatory role during osteogenesis. In this study, we inserted two important miRNAs, including miR-22 and miR-126 in the electrospun polycaprolactone (PCL) nanofibers and after scaffold characterization, osteoinductivity of the fabricated nanofibers was investigated by evaluating of the osteogenic differentiation potential of induced pluripotent stem cells (iPSCs) when grown on miRNAs-incorporated PCL nanofibers (PCL-miR) and empty PCL. MiRNAs incorporation had no effect on the fibers size and morphology, cell attachment, and protein adsorption, although viability and proliferation rate of the human iPSCs were increased after a week in PCL-miR compared to the empty PCL. The results obtained from alkaline phosphatase activity, calcium content, bone-related genes, and proteins expression assays demonstrated that the highest osteogenic markers were observed in iPSCs grown on the PCL-miR compared to the cells cultured on PCL and culture plate. According to the results, miR-incorporated PCL nanofibers could be considered as a promising potential tissue-engineered construct for the treatment of patients with bone lesions and defects.
Collapse
Affiliation(s)
- Aylin Tahmasebi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Seyed E Enderami
- Immunogenetics research center, Department of Medical Biotechnology, Faculty of Medicine, Mazandaran university of Medical Sciences, Sari, Iran
| | - Ehsan Saburi
- Medical Genetics and Molecular Medicine Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Islami
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Somayeh Yaslianifard
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Javad A Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Abdolreza Ardeshirylajimi
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biochemistry, Saint Louis University, St. Louis, Missouri
| | - Fatemeh Soleimanifar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Abbas S Moghadam
- Department of Immunogenetics, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
33
|
Zha F, Chen W, Zhang L, Yu D. Electrospun natural polymer and its composite nanofibrous scaffolds for nerve tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 31:519-548. [DOI: 10.1080/09205063.2019.1697170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Fangwen Zha
- Department of Chemistry, MOE Key Laboratory for Non-Equilibrium Synthesis and Modulation of Condensed Matter, School of Science, State Key Laboratory of Electrical Insulation and Power Equipments, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Wei Chen
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, PR China
| | - Lifeng Zhang
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, NC A&T State University, Greensboro, NC, USA
| | - Demei Yu
- Department of Chemistry, MOE Key Laboratory for Non-Equilibrium Synthesis and Modulation of Condensed Matter, School of Science, State Key Laboratory of Electrical Insulation and Power Equipments, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| |
Collapse
|
34
|
Farzamfar S, Salehi M, Tavangar SM, Verdi J, Mansouri K, Ai A, Malekshahi ZV, Ai J. A novel polycaprolactone/carbon nanofiber composite as a conductive neural guidance channel: an in vitro and in vivo study. Prog Biomater 2019; 8:239-248. [PMID: 31833033 PMCID: PMC6930318 DOI: 10.1007/s40204-019-00121-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/16/2019] [Indexed: 01/07/2023] Open
Abstract
The current study aimed to investigate the potential of carbon nanofibers to promote peripheral nerve regeneration. The carbon nanofiber-imbedded scaffolds were produced from polycaprolactone and carbon nanofibers using thermally induced phase separation method. Electrospinning technique was utilized to fabricate polycaprolactone/collagen nanofibrous sheets. The incorporation of carbon nanofibers into polycaprolactone's matrix significantly reduced its electrical resistance from 4.3 × 109 ± 0.34 × 109 Ω to 8.7 × 104 ± 1.2 × 104 Ω. Further in vitro studies showed that polycaprolactone/carbon nanofiber scaffolds had the porosity of 82.9 ± 3.7% and degradation rate of 1.84 ± 0.37% after 30 days and 3.58 ± 0.39% after 60 days. The fabricated scaffolds were favorable for PC-12 cells attachment and proliferation. Neural guidance channels were produced from the polycaprolactone/carbon nanofiber composites using water jet cutter machine then incorporated with PCL/collagen nanofibrous sheets. The composites were implanted into severed rat sciatic nerve. After 12 weeks, the results of histopathological examinations and functional analysis proved that conductive conduit out-performed the non-conductive type and induced no toxicity or immunogenic reactions, suggesting its potential applicability to treat peripheral nerve damage in the clinic.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Verdi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Korosh Mansouri
- Neuromusculoskletal Research Centre Firozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Arman Ai
- School of Medicine, Tehran University of Medical Sciences, Tehran, 141556447, Iran
| | - Ziba Veisi Malekshahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Wang Z, Mithieux SM, Weiss AS. Fabrication Techniques for Vascular and Vascularized Tissue Engineering. Adv Healthc Mater 2019; 8:e1900742. [PMID: 31402593 DOI: 10.1002/adhm.201900742] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Impaired or damaged blood vessels can occur at all levels in the hierarchy of vascular systems from large vasculatures such as arteries and veins to meso- and microvasculatures such as arterioles, venules, and capillary networks. Vascular tissue engineering has become a promising approach for fabricating small-diameter vascular grafts for occlusive arteries. Vascularized tissue engineering aims to fabricate meso- and microvasculatures for the prevascularization of engineered tissues and organs. The ideal small-diameter vascular graft is biocompatible, bridgeable, and mechanically robust to maintain patency while promoting tissue remodeling. The desirable fabricated meso- and microvasculatures should rapidly integrate with the host blood vessels and allow nutrient and waste exchange throughout the construct after implantation. A number of techniques used, including engineering-based and cell-based approaches, to fabricate these synthetic vasculatures are herein explored, as well as the techniques developed to fabricate hierarchical structures that comprise multiple levels of vasculature.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Suzanne M. Mithieux
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Anthony S. Weiss
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
- Bosch Institute University of Sydney NSW 2006 Australia
- Sydney Nano Institute University of Sydney NSW 2006 Australia
| |
Collapse
|
36
|
Scott KE, Rychel K, Ranamukhaarachchi S, Rangamani P, Fraley SI. Emerging themes and unifying concepts underlying cell behavior regulation by the pericellular space. Acta Biomater 2019; 96:81-98. [PMID: 31176842 DOI: 10.1016/j.actbio.2019.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 12/29/2022]
Abstract
Cells reside in a complex three-dimensional (3D) microenvironment where physical, chemical, and architectural features of the pericellular space regulate important cellular functions like migration, differentiation, and morphogenesis. A major goal of tissue engineering is to identify which properties of the pericellular space orchestrate these emergent cell behaviors and how. In this review, we highlight recent studies at the interface of biomaterials and single cell biophysics that are lending deeper insight towards this goal. Advanced methods have enabled the decoupling of architectural and mechanical features of the microenvironment, revealing multiple mechanisms of adhesion and mechanosensing modulation by biomaterials. Such studies are revealing important roles for pericellular space degradability, hydration, and adhesion competition in cell shape, volume, and differentiation regulation. STATEMENT OF SIGNIFICANCE: Cell fate and function are closely regulated by the local extracellular microenvironment. Advanced methods at the interface of single cell biophysics and biomaterials have shed new light on regulators of cell-pericellular space interactions by decoupling more features of the complex pericellular milieu than ever before. These findings lend deeper mechanistic insight into how biomaterials can be designed to fine-tune outcomes like differentiation, migration, and collective morphogenesis.
Collapse
Affiliation(s)
- Kiersten E Scott
- Bioengineering, University of California San Diego Jacobs School of Engineering, 9500 Gilman Drive #0435, La Jolla, CA 92093, USA.
| | - Kevin Rychel
- Bioengineering, University of California San Diego Jacobs School of Engineering, 9500 Gilman Drive #0435, La Jolla, CA 92093, USA.
| | - Sural Ranamukhaarachchi
- Bioengineering, University of California San Diego Jacobs School of Engineering, 9500 Gilman Drive #0435, La Jolla, CA 92093, USA.
| | - Padmini Rangamani
- Mechanical and Aerospace Engineering, University of California San Diego Jacobs School of Engineering, 9500 Gilman Drive #0411, La Jolla, CA 92093, USA.
| | - Stephanie I Fraley
- Bioengineering, University of California San Diego Jacobs School of Engineering, 9500 Gilman Drive #0435, La Jolla, CA 92093, USA.
| |
Collapse
|
37
|
Jahanmard F, Baghban Eslaminejad M, Amani-Tehran M, Zarei F, Rezaei N, Croes M, Amin Yavari S. Incorporation of F-MWCNTs into electrospun nanofibers regulates osteogenesis through stiffness and nanotopography. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 106:110163. [PMID: 31753334 DOI: 10.1016/j.msec.2019.110163] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/16/2019] [Accepted: 09/04/2019] [Indexed: 10/26/2022]
Abstract
Nanotopography and stiffness are major physical cues affecting cell fate. However, the current nanofiber modifications techniques are limited by their ability to control these two physical cues irrespective of each other without changing the materials' surface chemistry. For this reason, the isolated effects of topography and stiffness on osteogenic regulation in electrospun nanofibers have been studied incompletely. Here, we investigated 1. how functionalized multiwall carbon nanotubes (F-MWCNTs) loaded in Polycaprolactone (PCL) nanofibers control their physical properties and 2. whether the resulting unique structures lead to distinctive phenotypes in bone progenitor cells. Changes in material properties were measured by high-resolution electron microscopes, protein adsorption and tensile tests. The effect of the developed structures on human mesenchymal stem cell (MSC) osteogenic differentiation was determined by extensive quantification of early and late osteogenic marker genes. It was found that F-MWCNT loading was an effective method to independently control the PCL nanofiber surface nanoroughness or stiffness, depending on the applied F-MWCNT concentration. Collectively, this suggests that stiffness and topography activate distinct osteogenic signaling pathway. The current strategy can help our further understanding of the mechano-biological responses in osteoprogenitor cells, which could ultimately lead to improved design of bone substitute biomaterials.
Collapse
Affiliation(s)
- Fatemeh Jahanmard
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, Iran; Nanotechnology Institute, Amirkabir University of Technology, P.O. Box: 15875-4413, Tehran, Iran.
| | - Mohamadreza Baghban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, Iran.
| | - Mohammad Amani-Tehran
- Department of Textile Engineering, Amirkabir University of Technology, P.O. Box: 15875-4413, Tehran, Iran
| | - Fatemeh Zarei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, P.O. Box: 16635-148, Tehran, Iran
| | - Naeimeh Rezaei
- Department of Cell and Molecular Biology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Michiel Croes
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Centre Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, the Netherlands
| |
Collapse
|
38
|
Calejo I, Costa‐Almeida R, Reis RL, Gomes ME. A Textile Platform Using Continuous Aligned and Textured Composite Microfibers to Engineer Tendon-to-Bone Interface Gradient Scaffolds. Adv Healthc Mater 2019; 8:e1900200. [PMID: 31190369 DOI: 10.1002/adhm.201900200] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/20/2019] [Indexed: 12/30/2022]
Abstract
Tendon-to-bone interfaces exhibit a hierarchical multitissue transition. To replicate the progression from mineralized to nonmineralized tissue, a novel 3D fibrous scaffold is fabricated with spatial control over mineral distribution and cellular alignment. For this purpose, wet-spun continuous microfibers are produced using polycaprolactone (PCL)/ gelatin and PCL/gelatin/hydroxyapatite nano-to-microparticles (HAp). Higher extrusion rates result in aligned PCL/gelatin microfibers while, in the case of PCL/gelatin/HAp, the presence of minerals leads to a less organized structure. Biological performance using human adipose-derived stem cells (hASCs) demonstrates that topography of PCL/gelatin microfibers can induce cytoskeleton elongation, resembling native tenogenic organization. Matrix mineralization on PCL/gelatin/HAp wet-spun composite microfibers suggest the production of an osteogenic-like matrix, without external addition of osteogenic medium supplementation. As proof of concept, a 3D gradient structure is produced by assembling PCL/gelatin and PCL/gelatin/HAp microfibers, resulting in a fibrous scaffold with a continuous topographical and compositional gradient. Overall, the feasibility of wet-spinning for the generation of continuously aligned and textured microfibers is demonsrated, which can be further assembled into more complex 3D gradient structures to mimic characteristic features of tendon-to-bone interfaces.
Collapse
Affiliation(s)
- Isabel Calejo
- 3B's Research Groupi3Bs ‐ Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, Parque de Ciência e Tecnologia Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's‐ PT Government Associate Laboratory 4806‐909 Braga Guimarães Portugal
| | - Raquel Costa‐Almeida
- 3B's Research Groupi3Bs ‐ Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, Parque de Ciência e Tecnologia Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's‐ PT Government Associate Laboratory 4806‐909 Braga Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Groupi3Bs ‐ Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, Parque de Ciência e Tecnologia Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's‐ PT Government Associate Laboratory 4806‐909 Braga Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| | - Manuela E. Gomes
- 3B's Research Groupi3Bs ‐ Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAvePark, Parque de Ciência e Tecnologia Zona Industrial da Gandra 4805‐017 Barco Guimarães Portugal
- ICVS/3B's‐ PT Government Associate Laboratory 4806‐909 Braga Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineHeadquarters at University of Minho Avepark 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
39
|
Unal DB, Caliari SR, Lampe KJ. Engineering biomaterial microenvironments to promote myelination in the central nervous system. Brain Res Bull 2019; 152:159-174. [PMID: 31306690 DOI: 10.1016/j.brainresbull.2019.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023]
Abstract
Promoting remyelination and/or minimizing demyelination are key therapeutic strategies under investigation for diseases and injuries like multiple sclerosis (MS), spinal cord injury, stroke, and virus-induced encephalopathy. Myelination is essential for efficacious neuronal signaling. This myelination process is originated by oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). Resident OPCs are capable of both proliferation and differentiation, and also migration to demyelinated injury sites. OPCs can then engage with these unmyelinated or demyelinated axons and differentiate into myelin-forming oligodendrocytes (OLs). However this process is frequently incomplete and often does not occur at all. Biomaterial strategies can now be used to guide OPC and OL development with the goal of regenerating healthy myelin sheaths in formerly damaged CNS tissue. Growth and neurotrophic factors delivered from such materials can promote proliferation of OPCs or differentiation into OLs. While cell transplantation techniques have been used to replace damaged cells in wound sites, they have also resulted in poor transplant cell viability, uncontrollable differentiation, and poor integration into the host. Biomaterial scaffolds made from extracellular matrix (ECM) mimics that are naturally or synthetically derived can improve transplanted cell survival, support both transplanted and endogenous cell populations, and direct their fate. In particular, stiffness and degradability of these scaffolds are two parameters that can influence the fate of OPCs and OLs. The future outlook for biomaterials research includes 3D in vitro models of myelination / remyelination / demyelination to better mimic and study these processes. These models should provide simple relationships of myelination to microenvironmental biophysical and biochemical properties to inform improved therapeutic approaches.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States.
| |
Collapse
|
40
|
Petrova VA, Chernyakov DD, Poshina DN, Gofman IV, Romanov DP, Mishanin AI, Golovkin AS, Skorik YA. Electrospun Bilayer Chitosan/Hyaluronan Material and Its Compatibility with Mesenchymal Stem Cells. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2016. [PMID: 31238491 PMCID: PMC6631200 DOI: 10.3390/ma12122016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/17/2022]
Abstract
A bilayer nonwoven material for tissue regeneration was prepared from chitosan (CS) and hyaluronic acid (HA) by needleless electrospinning wherein 10-15 wt% (with respect to polysaccharide) polyethylene oxide was added as spinning starter. A fiber morphology study confirmed the material's uniform defect-free structure. The roughness of the bilayer material was in the range of 1.5-3 μm, which is favorable for cell growth. Electrospinning resulted in the higher orientation of the polymer structure compared with that of corresponding films, and this finding may be related to the orientation of the polymer chains during the spinning process. These structural changes increased the intermolecular interactions. Thus, despite a high swelling degree of 1.4-2.8 g/g, the bilayer matrix maintained its shape due to the large quantity of polyelectrolyte contacts between the chains of oppositely charged polymers. The porosity of the bilayer CS-HA nonwoven material was twice lower, while the Young's modulus and break stress were twice higher than that of a CS monolayer scaffold. Therefore, during the electrospinning of the second layer, HA may have penetrated into the pores of the CS layer, thereby increasing the polyelectrolyte contacts between the two polymers. The bilayer CS-HA scaffold exhibited good compatibility with mesenchymal stem cells. This characteristic makes the developed material promising for tissue engineering applications.
Collapse
Affiliation(s)
- Valentina A Petrova
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St Petersburg, Russia.
| | - Daniil D Chernyakov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St Petersburg, Russia.
| | - Daria N Poshina
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St Petersburg, Russia.
| | - Iosif V Gofman
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St Petersburg, Russia.
| | - Dmitry P Romanov
- Institute of Silicate Chemistry of the Russian Academy of Sciences, Adm. Makarova emb. 2, 199034 St. Petersburg, Russia.
| | - Alexander I Mishanin
- Almazov National Medical Research Centre, Akkuratova str. 2., 197341 St. Petersburg, Russia.
| | - Alexey S Golovkin
- Almazov National Medical Research Centre, Akkuratova str. 2., 197341 St. Petersburg, Russia.
| | - Yury A Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St Petersburg, Russia.
- Almazov National Medical Research Centre, Akkuratova str. 2., 197341 St. Petersburg, Russia.
| |
Collapse
|
41
|
Walker BW, Lara RP, Mogadam E, Yu CH, Kimball W, Annabi N. Rational Design of Microfabricated Electroconductive Hydrogels for Biomedical Applications. Prog Polym Sci 2019; 92:135-157. [PMID: 32831422 PMCID: PMC7441850 DOI: 10.1016/j.progpolymsci.2019.02.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Electroconductive hydrogels (ECHs) are highly hydrated 3D networks generated through the incorporation of conductive polymers, nanoparticles, and other conductive materials into polymeric hydrogels. ECHs combine several advantageous properties of inherently conductive materials with the highly tunable physical and biochemical properties of hydrogels. Recently, the development of biocompatible ECHs has been investigated for various biomedical applications, such as tissue engineering, drug delivery, biosensors, flexible electronics, and other implantable medical devices. Several methods for the synthesis of ECHs have been reported, which include the incorporation of electrically conductive materials such as gold and silver nanoparticles, graphene, and carbon nanotubes, as well as various conductive polymers (CPs), such as polyaniline, polypyrrole, and poly(3,4-ethylenedioxyythiophene) into hydrogel networks. Theses electroconductive composite hydrogels can be used as scaffolds with high swellability, tunable mechanical properties, and the capability to support cell growth both in vitro and in vivo. Furthermore, recent advancements in microfabrication techniques such as three dimensional (3D) bioprinting, micropatterning, and electrospinning have led to the development of ECHs with biomimetic microarchitectures that reproduce the characteristics of the native extracellular matrix (ECM). In addition, smart ECHs with controlled structures and healing properties have also been engineered into devices with prolonged half-lives and increased durability. The combination of sophisticated synthesis chemistries and modern microfabrication techniques have led to engineer smart ECHs with advanced architectures, geometries, and functionalities that are being increasingly used in drug delivery systems, biosensors, tissue engineering, and soft electronics. In this review, we will summarize different strategies to synthesize conductive biomaterials. We will also discuss the advanced microfabrication techniques used to fabricate ECHs with complex 3D architectures, as well as various biomedical applications of microfabricated ECHs.
Collapse
Affiliation(s)
- Brian W Walker
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Roberto Portillo Lara
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Zapopan, JAL, Mexico
| | - Emad Mogadam
- Department of Internal Medicine, Huntington Hospital, Pasadena, CA, 91105, USA
- Department of Internal Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Chu Hsiang Yu
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - William Kimball
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
42
|
Effect of Electrospun Fiber Mat Thickness and Support Method on Cell Morphology. NANOMATERIALS 2019; 9:nano9040644. [PMID: 31010029 PMCID: PMC6523829 DOI: 10.3390/nano9040644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
Abstract
Electrospun fiber mats (EFMs) are highly versatile biomaterials used in a myriad of biomedical applications. Whereas some facets of EFMs are well studied and can be highly tuned (e.g., pore size, fiber diameter, etc.), other features are under characterized. For example, although substrate mechanics have been explored by several groups, most studies rely on Young's modulus alone as a characterization variable. The influence of fiber mat thickness and the effect of supports are variables that are often not considered when evaluating cell-mechanical response. To assay the role of these features in EFM scaffold design and to improve understanding of scaffold mechanical properties, we designed EFM scaffolds with varying thickness (50-200 µm) and supporting methodologies. EFM scaffolds were comprised of polycaprolactone and were either electrospun directly onto a support, suspended across an annulus (3 or 10 mm inner diameter), or "tension-released" and then suspended across an annulus. Then, single cell spreading (i.e., Feret diameter) was measured in the presence of these different features. Cells were sensitive to EFM thickness and suspended gap diameter. Overall, cell spreading was greatest for 50 µm thick EFMs suspended over a 3 mm gap, which was the smallest thickness and gap investigated. These results are counterintuitive to conventional understanding in mechanobiology, which suggests that stiffer materials, such as thicker, supported EFMs, should elicit greater cell polarization. Additional experiments with 50 µm thick EFMs on polystyrene and polydimethylsiloxane (PDMS) supports demonstrated that cells can "feel" the support underlying the EFM if it is rigid, similar to previous results in hydrogels. These results also suggest that EFM curvature may play a role in cell response, separate from Young's modulus, possibly because of internal tension generated. These parameters are not often considered in EFM design and could improve scaffold performance and ultimately patient outcomes.
Collapse
|
43
|
Kumar D, Cain SA, Bosworth LA. Effect of Topography and Physical Stimulus on hMSC Phenotype Using a 3D In Vitro Model. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E522. [PMID: 30987078 PMCID: PMC6523693 DOI: 10.3390/nano9040522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/15/2019] [Accepted: 03/21/2019] [Indexed: 12/18/2022]
Abstract
This communication reports the first comparative study addressing the effects of both structural architecture and mechanical loading on human mesenchymal stem cells (hMSC) positioned at the interface of a 3D in vitro model composed of a nanofibre/hydrogel laminate composite. hMSC phenotype was affected by both stimuli over a seven-day period. Cells were orientated parallel to the underlying fibre direction irrespective of environment (electrospun 2D fibre sheet or laminate 2D sheet with collagen gel layer). Application of cyclical tensile force (5% strain, 1 Hz, 1 h per day) encouraged hMSCs to remain at the fibre/gel interface, whereas cells cultured in static conditions migrated from the interface to the upper hydrogel layer. Depending on the stimulus applied, hMSCs presented an up-regulation in gene expression, indicative of several cell lineages, with those cultured at the interface and physically stimulated expressing markers indicative of angiogenesis, osteogenesis, and tenogenesis. This study highlights the importance of developing biomaterial scaffolds with environmental cues to specifically drive cells towards the tissue intended for bioengineering.
Collapse
Affiliation(s)
- Deepak Kumar
- School of Materials, Faculty of Science and Engineering, University of Manchester, Manchester M13 9PL, UK.
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford, Oxford OX1 3QX, UK.
| | - Stuart A Cain
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Lucy A Bosworth
- School of Materials, Faculty of Science and Engineering, University of Manchester, Manchester M13 9PL, UK.
- Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK.
| |
Collapse
|
44
|
Tsai SW, Yu YL, Hsu FY. Fabrication of polycaprolactone tubular scaffolds with an orthogonal-bilayer structure for smooth muscle cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:308-314. [PMID: 30948066 DOI: 10.1016/j.msec.2019.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/30/2019] [Accepted: 03/04/2019] [Indexed: 01/06/2023]
Abstract
In this study, we used electrospinning to prepare a bilayered polycaprolactone (PCL) tubular graft consisting of an internal layer comprising axial nanofibers and an external layer comprising circumferentially aligned nanofibers. Subsequently, the surfaces of the electrospun PCL tubular scaffolds were modified with 1,6-diaminohexane to introduce amino groups and were then chemically conjugated with gelatin (Gel). The amino groups and Gel were successfully immobilized on the PCL scaffolds according to a ninhydrin assay, attenuated total reflectance Fourier transform infrared (ATR-FTIR) spectroscopic analysis and contact angle analysis. Additionally, vascular smooth muscle cells (vSMCs, A7r5) were cultured on random and aligned Gel-PCL scaffolds to evaluate the effects of fiber orientation on cell behavior. The results of immunofluorescence analysis showed that vSMCs on the aligned Gel-PCL scaffolds exhibited a pro-contractile phenotype.
Collapse
Affiliation(s)
- Shiao-Wen Tsai
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linko 33305, Taiwan; Department of Periodontics, Chang Gung Memorial Hospital, Taipei 10507, Taiwan
| | - Yen-Ling Yu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan
| | - Fu-Yin Hsu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan.
| |
Collapse
|
45
|
Mirzaei A, Saburi E, Islami M, Ardeshirylajimi A, Omrani MD, Taheri M, Moghadam AS, Ghafouri-Fard S. Bladder smooth muscle cell differentiation of the human induced pluripotent stem cells on electrospun Poly(lactide-co-glycolide) nanofibrous structure. Gene 2019; 694:26-32. [PMID: 30735717 DOI: 10.1016/j.gene.2019.01.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 01/01/2023]
Abstract
Smooth muscle cell (SMC) regeneration plays an important role in retrieving the bladder-wall functionality and it can be achieved by a proper cell-co-polymer constructed by tissue engineering. Human induced pluripotent stem cells (iPSCs), which can be specifically prepared for the patient, was considered as cells in this study, and Poly(lactide-co-glycolide) (PLGA) as a most interesting polymer in biomedical applications was applied to the scaffold fabrication by electrospinning. After scaffold characterization, SMC differentiation potential of the human iPSCs was investigated while cultured on the PLGA nanofibrous scaffold by evaluation of the SMC related important gene and protein markers. Alpha-smooth muscle actin (ASMA), Smooth muscle 22 alpha (SM-22a) as two early SMC markers were significantly up regulated either two and three weeks after differentiation induction in human iPSCs cultured on PLGA compared to those cells cultured on the tissue culture polystyrene (TCPS). But Calponin-1, Caldesmon1 and myosin heavy chain (MHC) expression differences in human iPSCs cultured on PLGA and TCPS were significant only three weeks after differentiation induction based on its lately expression in the differentiation process. ASMA and MHC proteins were also considered for evaluation by immunocytochemistry on differentiated iPSCs whereas results showed higher expression of these proteins in stem cells grown on PLGA compared to the TCPS. According to the results, human iPSCs demonstrated a great SMC differentiation potential when grown on PLGA and it could be considered as a promising cell-co-polymer for use in bladder tissue engineering.
Collapse
Affiliation(s)
- Ali Mirzaei
- Cellular & Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran; Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ehsan Saburi
- Immunogenetics and Cell Culture Department, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Islami
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Abdolreza Ardeshirylajimi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Shapouri Moghadam
- Bu-Ali Research Institute, Department of Immunogenetics, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Wu J, Zheng A, Liu Y, Jiao D, Zeng D, Wang X, Cao L, Jiang X. Enhanced bone regeneration of the silk fibroin electrospun scaffolds through the modification of the graphene oxide functionalized by BMP-2 peptide. Int J Nanomedicine 2019; 14:733-751. [PMID: 30705589 PMCID: PMC6342216 DOI: 10.2147/ijn.s187664] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Introduction Bone tissue engineering has become one of the most effective methods to treat bone defects. Silk fibroin (SF) is a natural protein with no physiological activities, which has features such as good biocompatibility and easy processing and causes minimal inflammatory reactions in the body. Scaffolds prepared by electrospinning SF can be used in bone tissue regeneration and repair. Graphene oxide (GO) is rich in functional groups, has good biocompatibility, and promotes osteogenic differentiation of stem cells, while bone morphogenetic protein-2 (BMP-2) polypeptide has an advantage in promoting osteogenesis induction. In this study, we attempted to graft BMP-2 polypeptide onto GO and then bonded the functionalized GO onto SF electrospun scaffolds through electrostatic interactions. The main purpose of this study was to further improve the biocompatibility of SF electrospun scaffolds, which could promote the osteogenic differentiation of bone marrow mesenchymal stem cells and the repair of bone tissue defects. Materials and methods The successful synthesis of GO and functionalized GO was confirmed by transmission electron microscope, X-ray photoelectron spectroscopy, and thermogravimetric analysis. Scanning electron microscopy, atomic force microscopy, mechanical test, and degradation experiment confirmed the preparation of SF electrospun scaffolds and the immobilization of GO on the fibers. In vitro experiment was used to verify the biocompatibility of the composite scaffolds, and in vivo experiment was used to prove the repairing ability of the composite scaffolds for bone defects. Results We successfully fabricated the composite scaffolds, which enhanced biocompatibility, not only promoting cell adhesion and proliferation but also greatly enhancing in vitro osteogenic differentiation of bone marrow stromal cells using either an osteogenic or non-osteogenic medium. Furthermore, transplantation of the composite scaffolds significantly promoted in vivo bone formation in critical-sized calvarial bone defects. Conclusion These findings suggested that the incorporation of BMP-2 polypeptide-functionalized GO into chitosan-coated SF electrospun scaffolds was a viable strategy for fabricating excellent scaffolds that enhance the regeneration of bone defects.
Collapse
Affiliation(s)
- Jiannan Wu
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| | - Ao Zheng
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| | - Yang Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Delong Jiao
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| | - Deliang Zeng
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| | - Xiao Wang
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| | - Lingyan Cao
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| | - Xinquan Jiang
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai JiaoTong University, School of Medicine, Shanghai 200011, China, ; .,Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200011, China, ;
| |
Collapse
|
47
|
Kong W, Qi Z, Xia P, Chang Y, Li H, Qu Y, Pan S, Yang X. Local delivery of FTY720 and NSCs on electrospun PLGA scaffolds improves functional recovery after spinal cord injury. RSC Adv 2019; 9:17801-17811. [PMID: 35520542 PMCID: PMC9064641 DOI: 10.1039/c9ra01717h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level. FTY720, also known as fingolimod, has recently been reported to exert a positive effect on the recovery from a spinal cord injury. Through local delivery to the lesion site, FTY720 effectively integrates with biomaterials, and the systemic adverse effects are alleviated. However, the effects of the proper mass ratio of FTY720 in biomaterials on neural stem cell (NSC) proliferation and differentiation, as well as functional recovery after SCI, have not been thoroughly investigated. In our study, we fabricated electrospun poly (lactide-co-glycolide) (PLGA)/FTY720 scaffolds at different mass ratios (0.1%, 1%, and 10%) and characterized these scaffolds. The effects of electrospun PLGA/FTY720 scaffolds on NSC proliferation and differentiation were measured. Then, a rat model of spinal transection was established to investigate the effects of PLGA/FTY720 scaffolds loaded with NSCs. Notably, 1% PLGA/FTY720 scaffolds exerted the best effects on the proliferation and differentiation of NSCs and 10% PLGA/FTY720 was cytotoxic to NSCs. Based on the Basso, Beattie, and Bresnahan (BBB) score, HE staining and immunofluorescence staining, the PLGA/FTY720 scaffold loaded with NSCs effectively promoted the recovery of spinal cord function. Thus, FTY720 properly integrated with electrospun PLGA scaffolds, and electrospun PLGA/FTY720 scaffolds loaded with NSCs may have potential applications for SCI as a nerve implant. Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level.![]()
Collapse
Affiliation(s)
- Weijian Kong
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Zhiping Qi
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Peng Xia
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yuxin Chang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Hongru Li
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yunpeng Qu
- Department of Cardiovascular Medicine
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Su Pan
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| |
Collapse
|
48
|
Nagam Hanumantharao S, Alinezhadbalalami N, Kannan S, Friske M, Rao S. Electrospun acellular scaffolds for mimicking the natural anisotropy of the extracellular matrix. RSC Adv 2019; 9:40190-40195. [PMID: 35542640 PMCID: PMC9076170 DOI: 10.1039/c9ra07777d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/28/2019] [Indexed: 01/05/2023] Open
Abstract
In tissue engineering, the use of scaffolds helps establish a synergistic relationship between the scaffolds and the tissues by improving cell–scaffold interaction. This interaction is enhanced when physiologically relevant biophysical cues are replicated in the artificial scaffolds. Here, we present a novel scaffold that mimics the natural anisotropy of the native extracellular matrix of tissues, fabricated by electrospinning a combination of three polymers: polycaprolactone (PCL), polyvinylidene fluoride (PVDF) and polyaniline (PANI). The scaffolds were characterized for their morphology, surface and mechanical properties. Rat cardiomyoblast (H9c2) cells, cultured on the PCL–PANI–PVDF scaffold, demonstrated cell alignment, penetration and proliferation across the entire surface area of the scaffold without any external chemical or physical stimuli. The PCL–PANI–PVDF scaffold, unlike other scaffolds, does not require post-processing or specific temperature conditions of storage, prior to use. These acellular scaffolds fabricated through polymer blending, open new avenues for research on functional acellular scaffolds for tissue engineering, based on synthetic materials. Co-axial electrospinning of three immiscible polymers by sequentially blending in solvents of different boiling points to obtain biomimetic scaffolds.![]()
Collapse
Affiliation(s)
| | | | - Srinivas Kannan
- Department of Biomedical Engineering
- Michigan Technological University
- Houghton
- USA
| | - Meghan Friske
- Department of Biomedical Engineering
- Michigan Technological University
- Houghton
- USA
| | - Smitha Rao
- Department of Biomedical Engineering
- Michigan Technological University
- Houghton
- USA
| |
Collapse
|
49
|
Hosseini FS, Soleimanifar F, Aidun A, Enderami SE, Saburi E, Marzouni HZ, Khani MM, Khojasteh A, Ardeshirylajimi A. Poly (3-hydroxybutyrate-co-3-hydroxyvalerate) improved osteogenic differentiation of the human induced pluripotent stem cells while considered as an artificial extracellular matrix. J Cell Physiol 2018; 234:11537-11544. [PMID: 30478907 DOI: 10.1002/jcp.27807] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/05/2018] [Indexed: 12/19/2022]
Abstract
Cocell polymers can be the best implants for replacing bone defects in patients. The pluripotent stem cells produced from the patient and the nanofibrous polymeric scaffold that can be completely degraded in the body and its produced monomers could be also usable are the best options for this implant. In this study, electrospun poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) nanofibers were fabricated and characterized and then osteogenic differentiation of the human-induced pluripotent stem cells (iPSCs) was investigated while cultured on PHBV scaffold. MTT results showed that cultured iPSCs on PHBV proliferation were increased compared to those cultured on tissue culture polystyrene (TCPS) as the control. Alkaline phosphatase (ALP) activity and calcium content were also significantly increased in iPSCs cultured on PHBV compared to the cultured on TCPS under osteogenic medium. Gene expression evaluation demonstrated that Runx2, collagen type I, ALP, osteonectin, and osteocalcin were upregulated in iPSCs cultured on PHBV scaffold in comparison with those cultured on TCPS for 2 weeks. Western blot analysis have shown that osteocalcin and osteopontin expression as two major osteogenic markers were increased in iPSCs cultured on PHBV scaffold. According to the results, nanofiber-based PHBV has a promising potential to increase osteogenic differentiation of the stem cells and iPSCs-PHBV as a cell-co-polymer construct demonstrated that has a great efficiency for use as a bone tissue engineered bioimplant.
Collapse
Affiliation(s)
- Fatemeh Sadat Hosseini
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Soleimanifar
- Dietary supplements and probiotic research center, Alborz University of Medical Sciences, Karaj, Iran
| | - Amir Aidun
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.,Tissues and Biomaterials Research Group (TBRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyedeh Elnaz Enderami
- Stem Cell and Regenerative Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering & Biotechnology (NIGEB), Tehran, Iran
| | - Ehsan Saburi
- Clinical Research Development Center, Imam Hasan Hospital, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hadi Zare Marzouni
- Department of Immunology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad-Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Khojasteh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Ardeshirylajimi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Development of a 3D Collagen Model for the In Vitro Evaluation of Magnetic-assisted Osteogenesis. Sci Rep 2018; 8:16270. [PMID: 30389949 PMCID: PMC6214996 DOI: 10.1038/s41598-018-33455-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
Magnetic stimulation has been applied to bone regeneration, however, the cellular and molecular mechanisms of repair still require a better understanding. A three-dimensional (3D) collagen model was developed using plastic compression, which produces dense, cellular, mechanically strong native collagen structures. Osteoblast cells (MG-63) and magnetic iron oxide nanoparticles (IONPs) were incorporated into collagen gels to produce a range of cell-laden models. A magnetic bio-reactor to support cell growth under static magnetic fields (SMFs) was designed and fabricated by 3D printing. The influences of SMFs on cell proliferation, differentiation, extracellular matrix production, mineralisation and gene expression were evaluated. Polymerase chain reaction (PCR) further determined the effects of SMFs on the expression of runt-related transcription factor 2 (Runx2), osteonectin (ON), and bone morphogenic proteins 2 and 4 (BMP-2 and BMP-4). Results demonstrate that SMFs, IONPs and the collagen matrix can stimulate the proliferation, alkaline phosphatase production and mineralisation of MG-63 cells, by influencing matrix/cell interactions and encouraging the expression of Runx2, ON, BMP-2 and BMP-4. Therefore, the collagen model developed here not only offers a novel 3D bone model to better understand the effect of magnetic stimulation on osteogenesis, but also paves the way for further applications in tissue engineering and regenerative medicine.
Collapse
|