1
|
Sandhoff CA, Loewen A, Kuhn Y, Vidal HT, Ruetten S, Jockenhoevel S. The Challenge of E-Spinning Sub-Millimeter Tubular Scaffolds-A Design-of-Experiments Study for Fiber Yield Improvement. Polymers (Basel) 2024; 16:1475. [PMID: 38891422 PMCID: PMC11174914 DOI: 10.3390/polym16111475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
In tissue engineering, electrospinning has gained significant interest due to its highly porous structure with an excellent surface area to volume ratio and fiber diameters that can mimic the structure of the extracellular matrix. Bioactive substances such as growth factors and drugs are easily integrated. In many applications, there is an important need for small tubular structures (I.D. < 1 mm). However, fabricating sub-millimeter structures is challenging as it reduces the collector area and increases the disturbing factors, leading to significant fiber loss. This study aims to establish a reliable and reproducible electrospinning process for sub-millimeter tubular structures with minimized material loss. Influencing factors were analyzed, and disturbance factors were removed before optimizing control variables through the design-of-experiments method. Structural and morphological characterization was performed, including the yield, thickness, and fiber arrangement of the scaffold. We evaluated the electrospinning process to enhance the manufacturing efficiency and reduce material loss. The results indicated that adjusting the voltage settings and polarity significantly increased the fiber yield from 8% to 94%. Variations in the process parameters also affected the scaffold thickness and homogeneity. The results demonstrate the complex relationship between the process parameters and provide valuable insights for optimizing electrospinning, particularly for the cost-effective and reproducible production of small tubular diameters.
Collapse
Affiliation(s)
- Cilia A. Sandhoff
- Department of Biohybrid & Medical Textiles (BioTex), AME Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany; (C.A.S.); (A.L.); (Y.K.)
| | - Alexander Loewen
- Department of Biohybrid & Medical Textiles (BioTex), AME Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany; (C.A.S.); (A.L.); (Y.K.)
| | - Yasmin Kuhn
- Department of Biohybrid & Medical Textiles (BioTex), AME Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany; (C.A.S.); (A.L.); (Y.K.)
| | - Haude-Tukua Vidal
- Laboratoire de Physique et Mécanique Textiles (LPMT), École Nationale Supérieure d’Ingénieurs Sud-Alsace, 12 Rue des Frères Lumière, 68 093 Mulhouse, France
| | - Stephan Ruetten
- Electron Microscopy Facility, University Hospital Aachen, Pauwelsstr. 30, 52074 Aachen, Germany;
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex), AME Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany; (C.A.S.); (A.L.); (Y.K.)
- Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Urmonderbaan 22, 6167 RD Geleen, The Netherlands
| |
Collapse
|
2
|
Xu X, Hu J, Xue H, Hu Y, Liu YN, Lin G, Liu L, Xu RA. Applications of human and bovine serum albumins in biomedical engineering: A review. Int J Biol Macromol 2023; 253:126914. [PMID: 37716666 DOI: 10.1016/j.ijbiomac.2023.126914] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Abstract
Serum albumin, commonly recognized as a predominant major plasma protein, is ubiquitously distributed among vertebrates, demonstrating versatility and widespread accessibility. Numerous studies have discussed the composition and attributes of human and bovine serum albumin; nonetheless, few systematic and comprehensive summaries on human and bovine serum albumin exist. This paper reviews the applications of human and bovine serum albumin in biomedical engineering. First, we introduce the differences in the structure of human and bovine serum albumin. Next, we describe the extraction methods for human and bovine serum albumin (fractionation process separation, magnetic adsorption, reverse micellar (RM) extraction, and genetic engineering) and the advantages and disadvantages of recently developed extraction methods. The characteristics of different processing forms of human and bovine serum albumin are also discussed, concomitantly elucidating their intrinsic properties, functions, and applications in biomedicine. Notably, their pivotal functions as carriers for drugs and tissue-engineered scaffolds, as well as their contributions to cell reproduction and bioimaging, are critically examined. Finally, to provide guidance for researchers in their future work, this review summarizes the current state of human and bovine serum albumin research and outlines potential future research topics.
Collapse
Affiliation(s)
- Xinhao Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jinyu Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Huaqian Xue
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; School of Pharmacy, Ningxia Medical University, Ningxia 750004, China
| | - Yingying Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ya-Nan Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Guanyang Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Ren-Ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
3
|
Gao H, Sun C, Shang S, Sun B, Sun M, Hu S, Yang H, Hu Y, Feng Z, Zhou W, Liu C, Wang J, Liu H. Wireless Electrical Signals Induce Functional Neuronal Differentiation of BMSCs on 3D Graphene Framework Driven by Magnetic Field. ACS NANO 2023; 17:16204-16220. [PMID: 37531596 DOI: 10.1021/acsnano.3c05725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are suggested as candidates for neurodegeneration therapy by autologous stem cells to overcome the lack of neural stem cells in adults. However, the differentiation of BMSCs into functional neurons is a major challenge for neurotherapy. Herein, a methodology has been proposed to induce functional neuronal differentiation of BMSCs on a conductive three-dimensional graphene framework (GFs) combined with a rotating magnetic field. A wireless electrical signal of about 10 μA can be generated on the surface of GFs by cutting the magnetic field lines based on the well-known electromagnetic induction effect, which has been proven to be suitable for inducing neuronal differentiation of BMSCs. The enhanced expressions of the specific genes/proteins and apparent Ca2+ intracellular flow indicate that BMSCs cultured on GFs with 15 min/day rotating magnetic field stimulation for 15 days can differentiate functional neurons without any neural inducing factor. The animal experiments confirm the neural differentiation of BMSCs on GFs after transplantation in vivo, accompanied by stimulation of an external rotating magnetic field. This study overcomes the lack of autologous neural stem cells for adult neurodegeneration patients and provides a facile and safe strategy to induce the neural differentiation of BMSCs, which has potential for clinical applications of neural tissue engineering.
Collapse
Affiliation(s)
- Haoyang Gao
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Chunhui Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Shuo Shang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Baojun Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Mingyuan Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Shuang Hu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Hongru Yang
- State Key Laboratory of Crystal Materials, Shandong University, 27 Shandanan Road, Jinan, Shandong 250100, People's Republic of China
| | - Ying Hu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Zhichao Feng
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Weijia Zhou
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Chao Liu
- Cryomedicine Laboratory, Qilu Hospital, Shandong University, Jinan 250012, People's Republic of China
| | - Jingang Wang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
| | - Hong Liu
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan 250022, People's Republic of China
- State Key Laboratory of Crystal Materials, Shandong University, 27 Shandanan Road, Jinan, Shandong 250100, People's Republic of China
| |
Collapse
|
4
|
Cao S, Bo R, Zhang Y. Polymeric Scaffolds for Regeneration of Central/Peripheral Nerves and Soft Connective Tissues. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Shunze Cao
- Applied Mechanics Laboratory Department of Engineering Mechanics Laboratory for Flexible Electronics Technology Tsinghua University Beijing 100084 China
| | - Renheng Bo
- Applied Mechanics Laboratory Department of Engineering Mechanics Laboratory for Flexible Electronics Technology Tsinghua University Beijing 100084 China
| | - Yihui Zhang
- Applied Mechanics Laboratory Department of Engineering Mechanics Laboratory for Flexible Electronics Technology Tsinghua University Beijing 100084 China
| |
Collapse
|
5
|
Visser Z, Verma SK, Rainey JK, Frampton JP. Loading and Release of Quercetin from Contact-Drawn Polyvinyl Alcohol Fiber Scaffolds. ACS Pharmacol Transl Sci 2022; 5:1305-1317. [PMID: 36524014 PMCID: PMC9745892 DOI: 10.1021/acsptsci.2c00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Indexed: 11/30/2022]
Abstract
Polymeric drug releasing systems have numerous applications for the treatment of chronic diseases and traumatic injuries. In this study, a simple, cost-effective, and scalable method for dry spinning of crosslinked polyvinyl alcohol (PVA) fibers is presented. This method utilizes an entangled solution of PVA to form liquid bridges that are drawn into rapidly drying fibers through extensional flow. The fibers are crosslinked by a one-pot reaction in which glyoxal is introduced to the PVA solution prior to contact drawing. Failure analysis of fiber formation is used to understand the interplay of polymer concentration, glyoxal concentration, and crosslinking time to identify appropriate formulations for the production of glyoxal-crosslinked PVA fibers. The small molecule quercetin (an anti-inflammatory plant flavonoid) can be added to the one-pot reaction and is shown to be incorporated into the fibers in a concentration-dependent manner. Upon rehydration in an aqueous medium, the glyoxal-crosslinked PVA fiber scaffolds retain their morphology and slowly degrade, as measured over the course of 10 days. As the scaffolds degrade, they release the loaded quercetin, reaching a cumulative release of 56 ± 6% of the loaded drug after 10 days. The bioactivity of the released quercetin is verified by combining quercetin-loaded fibers with contact-drawn polyethylene oxide-type I collagen (PEO-Col) fibers and monitoring the growth of PC12 cells on the fibers. PC12 cells readily attach to the PEO-Col fibers and display increased nerve growth factor-induced elongation and neurite formation in the presence of quercetin-loaded PVA fibers relative to substrates formed from only PEO-Col fibers or PEO-Col and PVA fibers without quercetin.
Collapse
Affiliation(s)
- Zachary
B. Visser
- School
of Biomedical Engineering, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
| | - Surendra Kumar Verma
- School
of Biomedical Engineering, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
| | - Jan K. Rainey
- School
of Biomedical Engineering, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
- Department
of Biochemistry & Molecular Biology, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
- Department
of Chemistry, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
| | - John P. Frampton
- School
of Biomedical Engineering, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
- Department
of Biochemistry & Molecular Biology, Dalhousie University, HalifaxB3H 4R2, Nova Scotia, Canada
| |
Collapse
|
6
|
Mahdipour E, Mequanint K. Films, Gels and Electrospun Fibers from Serum Albumin Globular Protein for Medical Device Coating, Biomolecule Delivery and Regenerative Engineering. Pharmaceutics 2022; 14:2306. [PMID: 36365125 PMCID: PMC9698923 DOI: 10.3390/pharmaceutics14112306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 09/18/2023] Open
Abstract
Albumin is a natural biomaterial that is abundantly available in blood and body fluids. It is clinically used as a plasma expander, thereby increasing the plasma thiol concentration due to its cysteine residues. Albumin is a regulator of intervascular oncotic pressure, serves as an anti-inflammatory modulator, and it has a buffering role due to its histidine imidazole residues. Because of its unique biological and physical properties, albumin has also emerged as a suitable biomaterial for coating implantable devices, for cell and drug delivery, and as a scaffold for tissue engineering and regenerative medicine. As a biomaterial, albumin can be used as surface-modifying film or processed either as cross-linked protein gels or as electrospun fibers. Herein we have discussed how albumin protein can be utilized in regenerative medicine as a hydrogel and as a fibrous mat for a diverse role in successfully delivering drugs, genes, and cells to targeted tissues and organs. The review of prior studies indicated that albumin is a tunable biomaterial from which different types of scaffolds with mechanical properties adjustable for various biomedical applications can be fabricated. Based on the progress made to date, we concluded that albumin-based device coatings, delivery of drugs, genes, and cells are promising strategies in regenerative and personalized medicine.
Collapse
Affiliation(s)
- Elahe Mahdipour
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
- Department of Medical Biotechnology & Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, University Ave., Mashhad 9177948564, Iran
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada
| |
Collapse
|
7
|
Kuten Pella O, Hornyák I, Horváthy D, Fodor E, Nehrer S, Lacza Z. Albumin as a Biomaterial and Therapeutic Agent in Regenerative Medicine. Int J Mol Sci 2022; 23:10557. [PMID: 36142472 PMCID: PMC9502107 DOI: 10.3390/ijms231810557] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 02/07/2023] Open
Abstract
Albumin is a constitutional plasma protein, with well-known biological functions, e.g., a nutrient for stem cells in culture. However, albumin is underutilized as a biomaterial in regenerative medicine. This review summarizes the advanced therapeutic uses of albumin, focusing on novel compositions that take advantage of the excellent regenerative potential of this protein. Albumin coating can be used for enhancing the biocompatibility of various types of implants, such as bone grafts or sutures. Albumin is mainly known as an anti-attachment protein; however, using it on implantable surfaces is just the opposite: it enhances stem cell adhesion and proliferation. The anticoagulant, antimicrobial and anti-inflammatory properties of albumin allow fine-tuning of the biological reaction to implantable tissue-engineering constructs. Another potential use is combining albumin with natural or synthetic materials that results in novel composites suitable for cardiac, neural, hard and soft tissue engineering. Recent advances in materials have made it possible to electrospin the globular albumin protein, opening up new possibilities for albumin-based scaffolds for cell therapy. Several described technologies have already entered the clinical phase, making good use of the excellent biological, but also regulatory, manufacturing and clinical features of serum albumin.
Collapse
Affiliation(s)
| | - István Hornyák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary
| | - Dénes Horváthy
- Department of Interventional Radiology, Semmelweis University, 1122 Budapest, Hungary
| | - Eszter Fodor
- Institute for Sports and Health Sciences, Hungarian University of Sports Science, 1123 Budapest, Hungary
| | - Stefan Nehrer
- Center for Regenerative Medicine, Danube University Krems, 3500 Krems an der Donau, Austria
| | - Zsombor Lacza
- Orthosera GmbH, 3500 Krems an der Donau, Austria
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary
- Institute for Sports and Health Sciences, Hungarian University of Sports Science, 1123 Budapest, Hungary
| |
Collapse
|
8
|
Tarricone G, Carmagnola I, Chiono V. Tissue-Engineered Models of the Human Brain: State-of-the-Art Analysis and Challenges. J Funct Biomater 2022; 13:146. [PMID: 36135581 PMCID: PMC9501967 DOI: 10.3390/jfb13030146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Neurological disorders affect billions of people across the world, making the discovery of effective treatments an important challenge. The evaluation of drug efficacy is further complicated because of the lack of in vitro models able to reproduce the complexity of the human brain structure and functions. Some limitations of 2D preclinical models of the human brain have been overcome by the use of 3D cultures such as cell spheroids, organoids and organs-on-chip. However, one of the most promising approaches for mimicking not only cell structure, but also brain architecture, is currently represented by tissue-engineered brain models. Both conventional (particularly electrospinning and salt leaching) and unconventional (particularly bioprinting) techniques have been exploited, making use of natural polymers or combinations between natural and synthetic polymers. Moreover, the use of induced pluripotent stem cells (iPSCs) has allowed the co-culture of different human brain cells (neurons, astrocytes, oligodendrocytes, microglia), helping towards approaching the central nervous system complexity. In this review article, we explain the importance of in vitro brain modeling, and present the main in vitro brain models developed to date, with a special focus on the most recent advancements in tissue-engineered brain models making use of iPSCs. Finally, we critically discuss achievements, main challenges and future perspectives.
Collapse
Affiliation(s)
- Giulia Tarricone
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
- Department of Chemistry and Industrial Chemistry, University of Genova, Via Dodecaneso 31, 16146 Genova, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| |
Collapse
|
9
|
Hsu CC, Serio A, Gopal S, Gelmi A, Chiappini C, Desai RA, Stevens MM. Biophysical Regulations of Epigenetic State and Notch Signaling in Neural Development Using Microgroove Substrates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32773-32787. [PMID: 35830496 PMCID: PMC9335410 DOI: 10.1021/acsami.2c01996] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A number of studies have recently shown how surface topography can alter the behavior and differentiation patterns of different types of stem cells. Although the exact mechanisms and molecular pathways involved remain unclear, a consistent portion of the literature points to epigenetic changes induced by nuclear remodeling. In this study, we investigate the behavior of clinically relevant neural populations derived from human pluripotent stem cells when cultured on polydimethylsiloxane microgrooves (3 and 10 μm depth grooves) to investigate what mechanisms are responsible for their differentiation capacity and functional behavior. Our results show that microgrooves enhance cell alignment, modify nuclear geometry, and significantly increase cellular stiffness, which we were able to measure at high resolution with a combination of light and electron microscopy, scanning ion conductance microscopy (SICM), and atomic force microscopy (AFM) coupled with quantitative image analysis. The microgrooves promoted significant changes in the epigenetic landscape, as revealed by the expression of key histone modification markers. The main behavioral change of neural stem cells on microgrooves was an increase of neuronal differentiation under basal conditions on the microgrooves. Through measurements of cleaved Notch1 levels, we found that microgrooves downregulate Notch signaling. We in fact propose that microgroove topography affects the differentiation potential of neural stem cells by indirectly altering Notch signaling through geometric segregation and that this mechanism in parallel with topography-dependent epigenetic modulations acts in concert to enhance stem cell neuronal differentiation.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Andrea Serio
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Sahana Gopal
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Amy Gelmi
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ciro Chiappini
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ravi A. Desai
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Molly M. Stevens
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| |
Collapse
|
10
|
An Engineered Protein-Based Building Block (Albumin Methacryloyl) for Fabrication of a 3D In Vitro Cryogel Model. Gels 2022; 8:gels8070404. [PMID: 35877489 PMCID: PMC9324498 DOI: 10.3390/gels8070404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of attrition in drug development or withdrawal; current animal experiments and traditional 2D cell culture systems fail to precisely predict the liver toxicity of drug candidates. Hence, there is an urgent need for an alternative in vitro model that can mimic the liver microenvironments and accurately detect human-specific drug hepatotoxicity. Here, for the first time we propose the fabrication of an albumin methacryloyl cryogel platform inspired by the liver’s microarchitecture via emulating the mechanical properties and extracellular matrix (ECM) cues of liver. Engineered crosslinkable albumin methacryloyl is used as a protein-based building block for fabrication of albumin cryogel in vitro models that can have potential applications in 3D cell culture and drug screening. In this work, protein modification, cryogelation, and liver ECM coating were employed to engineer highly porous three-dimensional cryogels with high interconnectivity, liver-like stiffness, and liver ECM as artificial liver constructs. The resulting albumin-based cryogel in vitro model provided improved cell–cell and cell–material interactions and consequently displayed excellent liver functional gene expression, being conducive to detection of fialuridine (FIAU) hepatotoxicity.
Collapse
|
11
|
Sharifi S, Saei AA, Gharibi H, Mahmoud NN, Harkins S, Dararatana N, Lisabeth EM, Serpooshan V, Végvári Á, Moore A, Mahmoudi M. Mass Spectrometry, Structural Analysis, and Anti-Inflammatory Properties of Photo-Cross-Linked Human Albumin Hydrogels. ACS APPLIED BIO MATERIALS 2022; 5:2643-2663. [PMID: 35544705 DOI: 10.1021/acsabm.2c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Albumin-based hydrogels offer unique benefits such as biodegradability and high binding affinity to various biomolecules, which make them suitable candidates for biomedical applications. Here, we report a non-immunogenic photocurable human serum-based (HSA) hydrogel synthesized by methacryloylation of human serum albumin by methacrylic anhydride (MAA). We used matrix-assisted laser desorption ionization-time-of-flight mass spectrometry, liquid chromatography-tandem mass spectrometry, as well as size exclusion chromatography to evaluate the extent of modification, hydrolytic and enzymatic degradation of methacrylated albumin macromer and its cross-linked hydrogels. The impacts of methacryloylation and cross-linking on alteration of inflammatory response and toxicity were evaluated in vitro using brain-derived HMC3 macrophages and Ex-Ovo chick chorioallantoic membrane assay. Results revealed that the lysines in HSA were the primary targets reacting with MAA, though modification of cysteine, threonine, serine, and tyrosine, with MAA was also confirmed. Both methacrylated HSA and its derived hydrogels were nontoxic and did not induce inflammatory pathways, while significantly reducing macrophage adhesion to the hydrogels; one of the key steps in the process of foreign body reaction to biomaterials. Cytokine and growth factor analysis showed that albumin-based hydrogels demonstrated anti-inflammatory response modulating cellular events in HMC3 macrophages. Ex-Ovo results also confirmed the biocompatibility of HSA macromer and hydrogels along with slight angiogenesis-modulating effects. Photocurable albumin hydrogels may be used as a non-immunogenic platform for various biomedical applications including passivation coatings.
Collapse
Affiliation(s)
- Shahriar Sharifi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Amir Ata Saei
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Hassan Gharibi
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | - Nouf N Mahmoud
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States.,Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.,Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar
| | - Shannon Harkins
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Naruphorn Dararatana
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Erika M Lisabeth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, United States.,Children's Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | - Ákos Végvári
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17 177 Stockholm, Sweden.,Proteomics Biomedicum, Division of Physiological Chemistry I, Department of Medical Biochemistry, Karolinska Institutet, SE-17 177 Stockholm, Sweden
| | - Anna Moore
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
12
|
Hsu CC, George JH, Waller S, Besnard C, Nagel DA, Hill EJ, Coleman MD, Korsunsky AM, Cui Z, Ye H. Increased connectivity of hiPSC-derived neural networks in multiphase granular hydrogel scaffolds. Bioact Mater 2022; 9:358-372. [PMID: 34820576 PMCID: PMC8586009 DOI: 10.1016/j.bioactmat.2021.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/17/2021] [Accepted: 07/07/2021] [Indexed: 12/21/2022] Open
Abstract
To reflect human development, it is critical to create a substrate that can support long-term cell survival, differentiation, and maturation. Hydrogels are promising materials for 3D cultures. However, a bulk structure consisting of dense polymer networks often leads to suboptimal microenvironments that impedes nutrient exchange and cell-to-cell interaction. Herein, granular hydrogel-based scaffolds were used to support 3D human induced pluripotent stem cell (hiPSC)-derived neural networks. A custom designed 3D printed toolset was developed to extrude hyaluronic acid hydrogel through a porous nylon fabric to generate hydrogel granules. Cells and hydrogel granules were combined using a weaker secondary gelation step, forming self-supporting cell laden scaffolds. At three and seven days, granular scaffolds supported higher cell viability compared to bulk hydrogels, whereas granular scaffolds supported more neurite bearing cells and longer neurite extensions (65.52 ± 11.59 μm) after seven days compared to bulk hydrogels (22.90 ± 4.70 μm). Long-term (three-month) cultures of clinically relevant hiPSC-derived neural cells in granular hydrogels supported well established neuronal and astrocytic colonies and a high level of neurite extension both inside and beyond the scaffold. This approach is significant as it provides a simple, rapid and efficient way to achieve a tissue-relevant granular structure within hydrogel cultures.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Julian H. George
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Sharlayne Waller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Cyril Besnard
- MBLEM, Department of Engineering Science, University of Oxford, Parks Road, Oxford, OX1 3PJ, UK
| | - David A Nagel
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
- Translational Medicine Research Group, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Eric J Hill
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Michael D. Coleman
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Alexander M. Korsunsky
- MBLEM, Department of Engineering Science, University of Oxford, Parks Road, Oxford, OX1 3PJ, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ, UK
| |
Collapse
|
13
|
Tyubaeva P, Varyan I, Krivandin A, Shatalova O, Karpova S, Lobanov A, Olkhov A, Popov A. The Comparison of Advanced Electrospun Materials Based on Poly(-3-hydroxybutyrate) with Natural and Synthetic Additives. J Funct Biomater 2022; 13:23. [PMID: 35323223 PMCID: PMC8955504 DOI: 10.3390/jfb13010023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/10/2022] Open
Abstract
The comparison of the effect of porphyrins of natural and synthetic origin containing the same metal atom on the structure and properties of the semi-crystalline polymer matrix is of current concern. A large number of modifying additives and biodegradable polymers for biomedical purposes, composed of poly(-3-hydroxybutyrate)-porphyrin, are of particular interest because of the combination of their unique properties. The objective of this work are electrospun fibrous material based on poly(-3-hydroxybutyrate) (PHB), hemin (Hmi), and tetraphenylporphyrin with iron (Fe(TPP)Cl). The structure of these new materials was investigated by methods such as optical and scanning electron microscopy, X-ray diffraction analysis, Electron paramagnetic resonance method, and Differential scanning calorimetry. The properties of the electrospun materials were analyzed by mechanical and biological tests, and the wetting contact angle was measured. In this work, it was found that even small concentrations of porphyrin can increase the antimicrobial properties by 12 times, improve the physical and mechanical properties by at least 3.5 times, and vary hydrophobicity by at least 5%. At the same time, additives similar in the structure had an oppositely directed effect on the supramolecular structure, the composition of the crystalline, and the amorphous phases. The article considers assumptions about the nature of such differences due to the influence of Hmi and Fe(TPP)Cl) on the macromolecular and fibrous structure of PHB.
Collapse
Affiliation(s)
- Polina Tyubaeva
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Per., 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Ivetta Varyan
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Per., 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Alexey Krivandin
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Olga Shatalova
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Svetlana Karpova
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Anton Lobanov
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Per., 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Anatoly Olkhov
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Per., 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| | - Anatoly Popov
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Per., 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Str., 119334 Moscow, Russia; (A.K.); (O.S.); (S.K.)
| |
Collapse
|
14
|
Tyubaeva P, Varyan I, Lobanov A, Olkhov A, Popov A. Effect of the Hemin Molecular Complexes on the Structure and Properties of the Composite Electrospun Materials Based on Poly(3-hydroxybutyrate). Polymers (Basel) 2021; 13:4024. [PMID: 34833324 PMCID: PMC8622405 DOI: 10.3390/polym13224024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
The creation of innovative fibrous materials based on biodegradable semicrystalline polymers and modifying additives is an urgent scientific problem. In particular, the development of biomedical materials based on molecular complexes and biopolymers with controlled properties is of great interest. The paper suggests an approach to modifying the structure and properties of the composite materials based on poly(3-hydroxybutyrate) (PHB) obtained by the electrospinning method using molecular complexes of hemin. The introduction of 1-5 wt. % of hemin has a significant effect on the supramolecular structure, morphology and properties of PHB-based fibers. Changes in the supramolecular structure intensified with the increasing hemin concentration. On the one hand, a decrease in the fraction of the crystalline phase by 8-10% was observed. At the same time, there is a decrease in the density of the amorphous phase by 15-70%. Moreover, the addition of hemin leads to an improvement in the strength characteristics of the material: the elongation at break increased by 1.5 times, and in the tensile strength, it increased by 3 times. The antimicrobial activity of the hemin-containing composite materials against Escherichia coli and Staphylococcus aureus was confirmed. The obtained materials are proposed to be used in the creation of composite systems for regenerative medicine.
Collapse
Affiliation(s)
- Polina Tyubaeva
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Lane, 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Street, 119334 Moscow, Russia
| | - Ivetta Varyan
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Lane, 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Street, 119334 Moscow, Russia
| | - Anton Lobanov
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Lane, 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Street, 119334 Moscow, Russia
| | - Anatoly Olkhov
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Lane, 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Street, 119334 Moscow, Russia
| | - Anatoly Popov
- Academic Department of Innovational Materials and Technologies Chemistry, Plekhanov Russian University of Economics, 36 Stremyanny Lane, 117997 Moscow, Russia; (I.V.); (A.L.); (A.O.); (A.P.)
- Department of Biological and Chemical Physics of Polymers, Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 4 Kosygina Street, 119334 Moscow, Russia
| |
Collapse
|
15
|
Su Y, Toftdal MS, Le Friec A, Dong M, Han X, Chen M. 3D Electrospun Synthetic Extracellular Matrix for Tissue Regeneration. SMALL SCIENCE 2021. [DOI: 10.1002/smsc.202100003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yingchun Su
- State Key Laboratory of Urban Water Resource and Environment School of Chemistry and Chemical Engineering Harbin Institute of Technology Harbin 150001 China
- Department of Biological and Chemical Engineering Aarhus University DK-8000 Aarhus C Denmark
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University DK-8000 Aarhus C Denmark
| | - Mette Steen Toftdal
- Department of Biological and Chemical Engineering Aarhus University DK-8000 Aarhus C Denmark
- Stem Cell Delivery and Pharmacology Novo Nordisk A/S DK-2760 Måløv Denmark
| | - Alice Le Friec
- Department of Biological and Chemical Engineering Aarhus University DK-8000 Aarhus C Denmark
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University DK-8000 Aarhus C Denmark
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment School of Chemistry and Chemical Engineering Harbin Institute of Technology Harbin 150001 China
| | - Menglin Chen
- Department of Biological and Chemical Engineering Aarhus University DK-8000 Aarhus C Denmark
- Interdisciplinary Nanoscience Center (iNANO) Aarhus University DK-8000 Aarhus C Denmark
| |
Collapse
|
16
|
Chen T, Jiang H, Zhu Y, Chen X, Zhang D, Li X, Shen F, Xia H, Min Y, Xie K. Highly Ordered 3D Tissue Engineering Scaffolds as a Versatile Culture Platform for Nerve Cells Growth. Macromol Biosci 2021; 21:e2100047. [PMID: 33893711 DOI: 10.1002/mabi.202100047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/11/2021] [Indexed: 12/14/2022]
Abstract
Tissue engineering scaffolds provide an encouraging alternative for nerve injuries due to their biological support for nerve cell growth, which can be used for neuronal repair. Nerve cells have been reported to be mostly cultured on 2D scaffolds that cannot mimic the native extracellular matrix. Herein, highly ordered 3D scaffolds are fabricated for nerve cell culture by melt electrospinning writing, the microstructures and geometries of the scaffolds could be well modulated. An effective strategy for scaffold surface modification to promote nerve cell growth is proposed. The effects of scaffolds with different surface modifications, viz., plasma treatment, single poly-D-lysine (PDL) coating after plasma treatment, single laminin (LM) coating after plasma treatment, double PDL and LM coatings after plasma treatment, on PC12 cell growth are evaluated. Experiments show the scaffold modified with double PDL and LM coatings after plasma treatment facilitated the growth of PC12 cells most effectively, indicating the synergistic effect of PDL and LM on the growth of nerve cells. This is the first systematic and quantitative study of the effects of different scaffold surface modifications on nerve cell growth. The above results provide a versatile culture platform for growing nerve cells, and for recovery from peripheral nerve injury.
Collapse
Affiliation(s)
- Tingkuo Chen
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Haiming Jiang
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yibin Zhu
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xueliu Chen
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Dao Zhang
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xiang Li
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Fangcheng Shen
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Hongyan Xia
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yonggang Min
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, 510006, China
| | - Kang Xie
- Dongyuan Synergy Innovation Institute for Modern Industries of GDUT, Guangdong University of Technology, Guangzhou, 510006, China
| |
Collapse
|
17
|
Casella A, Panitch A, Leach JK. Endogenous Electric Signaling as a Blueprint for Conductive Materials in Tissue Engineering. Bioelectricity 2021; 3:27-41. [PMID: 34476376 PMCID: PMC8370482 DOI: 10.1089/bioe.2020.0027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bioelectricity plays an important role in cell behavior and tissue modulation, but is understudied in tissue engineering research. Endogenous electrical signaling arises from the transmembrane potential inherent to all cells and contributes to many cell behaviors, including migration, adhesion, proliferation, and differentiation. Electrical signals are also involved in tissue development and repair. Synthetic and natural conductive materials are under investigation for leveraging endogenous electrical signaling cues in tissue engineering applications due to their ability to direct cell differentiation, aid in maturing electroactive cell types, and promote tissue functionality. In this review, we provide a brief overview of bioelectricity and its impact on cell behavior, report recent literature using conductive materials for tissue engineering, and discuss opportunities within the field to improve experimental design when using conductive substrates.
Collapse
Affiliation(s)
- Alena Casella
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
- Department of Surgery and UC Davis Health, Sacramento, California, USA
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| |
Collapse
|
18
|
Lutzweiler G, Barthes J, Charles AL, Ball V, Louis B, Geny B, Vrana NE. Improving the colonization and functions of Wharton's Jelly-derived mesenchymal stem cells by a synergetic combination of porous polyurethane scaffold with an albumin-derived hydrogel. ACTA ACUST UNITED AC 2020; 16:015005. [PMID: 33300500 DOI: 10.1088/1748-605x/abaf05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The development of neo-tissues assisted by artificial scaffolds is continually progressing, but the reproduction of the extracellular environment surrounding cells is quite complex. While synthetic scaffolds can support cell growth, they lack biochemical cues that can prompt cell proliferation or differentiation. In this study, Wharton's Jelly-derived mesenchymal stem cells are seeded on a polyurethane (PU) scaffold combined with a hydrogel based on bovine serum albumin (BSA). BSA hydrogel is obtained through thermal treatment. While such treatment leads to partial unfolding of the protein, we show that the extent of denaturation is small enough to maintain its bioactivity, such as protein binding. Therefore, BSA provides a suitable playground for cells inside the scaffold, allowing higher spreading, proliferation and matrix secretions. Furthermore, the poor mechanical properties of the hydrogel are compensated for by the porous PU scaffold, whose architecture is well controlled. We show that even though PU by itself can allow cell adhesion and protein secretion, cell proliferation is 3.5 times higher in the PU + BSA scaffolds as compared to pure PU after 21 d, along with the non-collagenous protein secretions (389 versus 134 μmmg -1). Conversely, the secretion of sulphated glycosaminoglycans is 12.3-fold higher in the scaffold made solely of PU. Thereby, we propose a simple approach to generating a hybrid material composed of a combination of PU and BSA hydrogel as a promising scaffold for tissue regeneration.
Collapse
Affiliation(s)
- G Lutzweiler
- Institut National de la Santé et de la Recherche Medicale, UMR_S 1121, 11 rue Humann, 67085, Strasbourg Cedex, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Mehdipour M, Mehdipour T, Skinner CM, Wong N, Liu C, Chen CC, Jeon OH, Zuo Y, Conboy MJ, Conboy IM. Plasma dilution improves cognition and attenuates neuroinflammation in old mice. GeroScience 2020; 43:1-18. [PMID: 33191466 PMCID: PMC8050203 DOI: 10.1007/s11357-020-00297-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023] Open
Abstract
Our recent study has established that young blood factors are not causal, nor necessary, for the systemic rejuvenation of mammalian tissues. Instead, a procedure referred to as neutral blood exchange (NBE) that resets signaling milieu to a pro-regenerative state through dilution of old plasma, enhanced the health and repair of the muscle and liver, and promoted better hippocampal neurogenesis in 2-year-old mice (Mehdipour et al., Aging 12:8790–8819, 2020). Here we expand the rejuvenative phenotypes of NBE, focusing on the brain. Namely, our results demonstrate that old mice perform much better in novel object and novel texture (whisker discrimination) tests after a single NBE, which is accompanied by reduced neuroinflammation (less-activated CD68+ microglia). Evidence against attenuation/dilution of peripheral senescence-associated secretory phenotype (SASP) as the main mechanism behind NBE was that the senolytic ABT 263 had limited effects on neuroinflammation and did not enhance hippocampal neurogenesis in the old mice. Interestingly, peripherally acting ABT 263 and NBE both diminished SA-βGal signal in the old brain, demonstrating that peripheral senescence propagates to the brain, but NBE was more robustly rejuvenative than ABT 263, suggesting that rejuvenation was not simply by reducing senescence. Explaining the mechanism of the positive effects of NBE on the brain, our comparative proteomics analysis demonstrated that dilution of old blood plasma yields an increase in the determinants of brain maintenance and repair in mice and in people. These findings confirm the paradigm of rejuvenation through dilution of age-elevated systemic factors and extrapolate it to brain health and function.
Collapse
Affiliation(s)
- Melod Mehdipour
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Taha Mehdipour
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Colin M Skinner
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Nathan Wong
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Chao Liu
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Chia-Chien Chen
- Department of Molecular and Cellular Biology and QB3, UCSC, Santa Cruz, CA, USA
| | - Ok Hee Jeon
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, USA.,Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yi Zuo
- Department of Molecular and Cellular Biology and QB3, UCSC, Santa Cruz, CA, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA, USA.
| |
Collapse
|
20
|
Ritzau-Reid KI, Spicer CD, Gelmi A, Grigsby CL, Ponder JF, Bemmer V, Creamer A, Vilar R, Serio A, Stevens MM. An Electroactive Oligo-EDOT Platform for Neural Tissue Engineering. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2003710. [PMID: 34035794 PMCID: PMC7610826 DOI: 10.1002/adfm.202003710] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 05/04/2023]
Abstract
The unique electrochemical properties of the conductive polymer poly(3,4-ethylenedioxythiophene):polystyrene sulfonate (PEDOT:PSS) make it an attractive material for use in neural tissue engineering applications. However, inadequate mechanical properties, and difficulties in processing and lack of biodegradability have hindered progress in this field. Here, the functionality of PEDOT:PSS for neural tissue engineering is improved by incorporating 3,4-ethylenedioxythiophene (EDOT) oligomers, synthesized using a novel end-capping strategy, into block co-polymers. By exploiting end-functionalized oligoEDOT constructs as macroinitiators for the polymerization of poly(caprolactone), a block co-polymer is produced that is electroactive, processable, and bio-compatible. By combining these properties, electroactive fibrous mats are produced for neuronal culture via solution electrospinning and melt electrospinning writing. Importantly, it is also shown that neurite length and branching of neural stem cells can be enhanced on the materials under electrical stimulation, demonstrating the promise of these scaffolds for neural tissue engineering.
Collapse
Affiliation(s)
- Kaja I. Ritzau-Reid
- Department of Materials, Department of Bioengineering, Institute of
Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Christopher D. Spicer
- Department of Materials, Department of Bioengineering, Institute of
Biomedical Engineering, Imperial College London, London SW7 2AZ, UK;
Department of Medical Biochemistry and Biophysics, Karolinska Institutet,
Stockholm 171 77, Sweden; Department of Chemistry, York Biomedical Research
Institute, University of York, Heslington YO10 5DD, UK
| | - Amy Gelmi
- Department of Materials, Department of Bioengineering, Institute of
Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Applied
Chemistry and Environmental Science, School of Science, RMIT University,
Melbourne 3000, Australia
| | - Christopher L. Grigsby
- Department of Medical Biochemistry and Biophysics, Karolinska
Institutet, Stockholm 171 77, Sweden
| | - James F. Ponder
- Department of Chemistry, Imperial College London, London SW7 2AZ,
UK
| | - Victoria Bemmer
- Department of Materials, Department of Bioengineering, Institute of
Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Adam Creamer
- Department of Materials, Department of Bioengineering, Institute of
Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Ramon Vilar
- Department of Chemistry, Imperial College London, London SW7 2AZ,
UK
| | - Andrea Serio
- Department of Materials, Department of Bioengineering, Institute of
Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Centre
for Craniofacial & Regenerative Biology, King’s College London
and The Francis Crick Institute, Tissue Engineering and Biophotonics
Division, Dental Institute, King’s College London, London SE1 9RT,
UK
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering, Institute
of Biomedical Engineering, Imperial College London, London SW7 2AZ, UK;
Department of Medical Biochemistry and Biophysics, Karolinska Institutet,
Stockholm 171 77, Sweden
| |
Collapse
|
21
|
Hájovská P, Chytil M, Kalina M. Rheological study of albumin and hyaluronan-albumin hydrogels: Effect of concentration, ionic strength, pH and molecular weight. Int J Biol Macromol 2020; 161:738-745. [DOI: 10.1016/j.ijbiomac.2020.06.063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/01/2020] [Accepted: 06/06/2020] [Indexed: 12/13/2022]
|
22
|
Zhu Y, Wang D, Yao X, Wang M, Zhao Y, Lu Y, Wang Z, Guo Y. Biomimetic hybrid scaffold of electrospun silk fibroin and pancreatic decellularized extracellular matrix for islet survival. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:151-165. [PMID: 32867627 DOI: 10.1080/09205063.2020.1818018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Islet transplantation is considered as one of the promising treatment options for curing diabetes. However, the extracellular matrix (ECM) is destroyed during the process of islet isolation and extraction, which leads to decreased islet activity in vitro. ECM-based biomaterials which used to reconstruct the microenvironment of cells have been applied in various fields. In this study, an electrospinning hybrid scaffolds with silk fibroin (SF) and pig pancreatic decellularized extracellular matrix (P-dECM) have been prepared to mimic the islet ECM in vivo. Furthermore, the activity and function of islet were evaluated in vitro. The microstructures, hydrophilia and the main components of scaffolds were characterized by SEM, contact angle analysis and immunohistochemical experiment. The toxicity of stents was assessed by MTT assay. Cell activity and function were estimated by the live-dead cell staining, immunofluorescence, glucose-stimulated insulin secretion assay and q-PCR. A nanofiber scaffold with good hydrophilicity, non-toxic and retention of key ECM components has been obtained, which can improve the survival and promote and function of islets. This scaffold can be a promising candidate for pancreatic tissue engineering and provides a new strategy for islet transplantation.
Collapse
Affiliation(s)
- Yi Zhu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China
| | - Dongzhi Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China
| | - Xihao Yao
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China
| | - Mingming Wang
- Medical School of Nantong University, Nantong, Jiangsu, P.R China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuhua Lu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China
| | - Zhiwei Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R China
| |
Collapse
|
23
|
Saeki K, Hiramatsu H, Hori A, Hirai Y, Yamada M, Utoh R, Seki M. Sacrificial Alginate-Assisted Microfluidic Engineering of Cell-Supportive Protein Microfibers for Hydrogel-Based Cell Encapsulation. ACS OMEGA 2020; 5:21641-21650. [PMID: 32905425 PMCID: PMC7469388 DOI: 10.1021/acsomega.0c02385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/04/2020] [Indexed: 05/04/2023]
Abstract
Although many types of technologies for hydrogel-based cell cultivation have recently been developed, strategies to integrate cell-adhesive micrometer-sized supports with bulk-scale hydrogel platforms have not been fully established. Here, we present a highly unique approach to produce cell-adhesive, protein-based microfibers assisted by the sacrificial template of alginate; we applied these fibers as microengineered scaffolds for hydrogel-based cell encapsulation. Two types of microfluidic devices were designed and fabricated: a single-layered device for producing relatively thick (Φ of 10-60 μm) alginate-protein composite fibers with a uniform cross-sectional morphology and a four-layered device for preparing thinner (Φ of ∼4 μm) ones through the formation of patterned microfibers with eight distinct alginate-protein composite regions. Following chemical cross-linking of protein molecules and the subsequent removal of the sacrificial alginate from the double-network matrices, microfibers composed only of cross-linked proteins were obtained. We used gelatin, albumin, and hemoglobin as the protein material, and the gelatin-based cell-adhesive fibers were further encapsulated in hydrogels together with the mammalian cells. We clarified that the thinner fibers were especially effective in promoting cell proliferation, and the shape of the constructs was maintained even after removing the hydrogel matrices. The presented approach offers cells with biocompatible solid supports that enhance cell adhesion and proliferation, paving the way for the next generation of techniques for tissue engineering and multicellular organoid formation.
Collapse
Affiliation(s)
- Kotone Saeki
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Hisataka Hiramatsu
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Ayaka Hori
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Yu Hirai
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Rie Utoh
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Minoru Seki
- Department of Applied Chemistry and
Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
24
|
Liu Z, Ramakrishna S, Liu X. Electrospinning and emerging healthcare and medicine possibilities. APL Bioeng 2020; 4:030901. [PMID: 32695956 PMCID: PMC7365682 DOI: 10.1063/5.0012309] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/01/2020] [Indexed: 01/06/2023] Open
Abstract
Electrospinning forms fibers from either an electrically charged polymer solution or polymer melt. Over the past decades, it has become a simple and versatile method for nanofiber production. Hence, it has been explored in many different applications. Commonly used electrospinning assembles fibers from polymer solutions in various solvents, known as solution electrospinning, while melt and near-field electrospinning techniques enhance the versatility of electrospinning. Adaption of additive manufacturing strategy to electrospinning permits precise fiber deposition and predefining pattern construction. This manuscript critically presents the potential of electrospun nanofibers in healthcare applications. Research community drew impetus from the similarity of electrospun nanofibers to the morphology and mechanical properties of fibrous extracellular matrices (ECM) of natural human tissues. Electrospun nanofibrous scaffolds act as ECM analogs for specific tissue cells, stem cells, and tumor cells to realize tissue regeneration, stem cell differentiation, and in vitro tumor model construction. The large surface-to-volume ratio of electrospun nanofibers offers a considerable number of bioactive agents binding sites, which makes it a promising candidate for a number of biomedical applications. The applications of electrospinning in regenerative medicine, tissue engineering, controlled drug delivery, biosensors, and cancer diagnosis are elaborated. Electrospun nanofiber incorporations in medical device coating, in vitro 3D cancer model, and filtration membrane are also discussed.
Collapse
Affiliation(s)
- Ziqian Liu
- Department of Mechanical, Materials and Manufacturing, The University of Nottingham Ningbo China, Ningbo 315100, China
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology, Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
| | - Xiaoling Liu
- Department of Mechanical, Materials and Manufacturing, The University of Nottingham Ningbo China, Ningbo 315100, China
| |
Collapse
|
25
|
A single-cell Raman-based platform to identify developmental stages of human pluripotent stem cell-derived neurons. Proc Natl Acad Sci U S A 2020; 117:18412-18423. [PMID: 32694205 PMCID: PMC7414136 DOI: 10.1073/pnas.2001906117] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We developed a label-free and noninvasive single-cell Raman microspectroscopy (SCRM)-based platform to identify neural cell lineages derived from clinically relevant human induced pluripotent stem cells (hiPSCs). Through large-scale Raman spectral analysis, we can distinguish hiPSCs and hiPSC-derived neural cells using their intrinsic biochemical profile. We identified glycogen as a Raman biomarker for neuronal differentiation and validated the results using conventional glycogen detection assays. The parameters obtained from SCRM were processed by a novel machine learning method based on t-distributed stochastic neighbor embedding (t-SNE)-enhanced ensemble stacking, enabling highly accurate and robust cell classification. The platform and the proposed biomarker should also be applicable to other cell types and can shed light on developmental biology and glycogen metabolism disorders. Stem cells with the capability to self-renew and differentiate into multiple cell derivatives provide platforms for drug screening and promising treatment options for a wide variety of neural diseases. Nevertheless, clinical applications of stem cells have been hindered partly owing to a lack of standardized techniques to characterize cell molecular profiles noninvasively and comprehensively. Here, we demonstrate that a label-free and noninvasive single-cell Raman microspectroscopy (SCRM) platform was able to identify neural cell lineages derived from clinically relevant human induced pluripotent stem cells (hiPSCs). By analyzing the intrinsic biochemical profiles of single cells at a large scale (8,774 Raman spectra in total), iPSCs and iPSC-derived neural cells can be distinguished by their intrinsic phenotypic Raman spectra. We identified a Raman biomarker from glycogen to distinguish iPSCs from their neural derivatives, and the result was verified by the conventional glycogen detection assays. Further analysis with a machine learning classification model, utilizing t-distributed stochastic neighbor embedding (t-SNE)-enhanced ensemble stacking, clearly categorized hiPSCs in different developmental stages with 97.5% accuracy. The present study demonstrates the capability of the SCRM-based platform to monitor cell development using high content screening with a noninvasive and label-free approach. This platform as well as our identified biomarker could be extensible to other cell types and can potentially have a high impact on neural stem cell therapy.
Collapse
|
26
|
Nguyen LTB, Hsu CC, Ye H, Cui Z. Development of an in situ injectable hydrogel containing hyaluronic acid for neural regeneration. ACTA ACUST UNITED AC 2020; 15:055005. [PMID: 32324167 DOI: 10.1088/1748-605x/ab8c43] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In this work, a novel enzymatically crosslinked injectable hydrogel comprising hyaluronic acid (HyA), dopamine (DA), and 3-(4-hydroxyphenyl) propionic acid (HPA) conjugates was successfully developed. To the best of our knowledge, it is the first time that HPA is conjugated to a HyA-based backbone. In situ hydrogelation of HyA-DA-HPA occurred in the presence of hydrogen peroxide (H2O2) as an oxidant and horseradish peroxidase (HRP) as a catalyst. Proton nuclear magnetic resonance and Fourier transform infrared spectroscopy were used to characterize the chemical reactions between HyA, DA, and HPA. Gel formation completed between 3 s to 5 min depending on the concentrations of polymer, HRP, and H2O2. Crosslinked HyA-DA-HPA gels acquired storage moduli ranging from ∼100 Pa to ∼20 000 Pa (at f = 2000 rad s-1). Biocompatibility of the hydrogels was examined with human mesenchymal stem cells (hMSCs) and human induced pluripotent stem cell-derived neural stem cells. The hydrogels made of 2.0 w/v% HyA-DA-HPA hydrogels, 0.24 U ml-1 HRP and ≤ 0.5 µmol ml-1 H2O2 were found biocompatible with hMSCs cultured on and encapsulated within the hydrogels. Since HyA serves as a backbone of the extracellular matrix in the central nervous system (CNS) and DA acquires the ability to restore dopaminergic neurons, use of this injectable HyA-DA-HPA hydrogel for stem cell transplantation is a potential treatment strategy for CNS repair and regeneration.
Collapse
Affiliation(s)
- Linh T B Nguyen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX3 7DQ, United Kingdom. Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London WC1X 8LD, United Kingdom
| | | | | | | |
Collapse
|
27
|
Mehdipour M, Skinner C, Wong N, Lieb M, Liu C, Etienne J, Kato C, Kiprov D, Conboy MJ, Conboy IM. Rejuvenation of three germ layers tissues by exchanging old blood plasma with saline-albumin. Aging (Albany NY) 2020; 12:8790-8819. [PMID: 32474458 PMCID: PMC7288913 DOI: 10.18632/aging.103418] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022]
Abstract
Heterochronic blood sharing rejuvenates old tissues, and most of the studies on how this works focus on young plasma, its fractions, and a few youthful systemic candidates. However, it was not formally established that young blood is necessary for this multi-tissue rejuvenation. Here, using our recently developed small animal blood exchange process, we replaced half of the plasma in mice with saline containing 5% albumin (terming it a "neutral" age blood exchange, NBE) thus diluting the plasma factors and replenishing the albumin that would be diminished if only saline was used. Our data demonstrate that a single NBE suffices to meet or exceed the rejuvenative effects of enhancing muscle repair, reducing liver adiposity and fibrosis, and increasing hippocampal neurogenesis in old mice, all the key outcomes seen after blood heterochronicity. Comparative proteomic analysis on serum from NBE, and from a similar human clinical procedure of therapeutic plasma exchange (TPE), revealed a molecular re-setting of the systemic signaling milieu, interestingly, elevating the levels of some proteins, which broadly coordinate tissue maintenance and repair and promote immune responses. Moreover, a single TPE yielded functional blood rejuvenation, abrogating the typical old serum inhibition of progenitor cell proliferation. Ectopically added albumin does not seem to be the sole determinant of such rejuvenation, and levels of albumin do not decrease with age nor are increased by NBE/TPE. A model of action (supported by a large body of published data) is that significant dilution of autoregulatory proteins that crosstalk to multiple signaling pathways (with their own feedback loops) would, through changes in gene expression, have long-lasting molecular and functional effects that are consistent with our observations. This work improves our understanding of the systemic paradigms of multi-tissue rejuvenation and suggest a novel and immediate use of the FDA approved TPE for improving the health and resilience of older people.
Collapse
Affiliation(s)
- Melod Mehdipour
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Colin Skinner
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Nathan Wong
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Michael Lieb
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Chao Liu
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Jessy Etienne
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Cameron Kato
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Dobri Kiprov
- California Pacific Medical Center, Apheresis Care Group, San-Francisco, CA 94115, USA
| | - Michael J. Conboy
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| | - Irina M. Conboy
- Department of Bioengineering and QB3, UC Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
28
|
Preparation of Multiwall Carbon Nanotubes Embedded Electroconductive Multi-Microchannel Scaffolds for Neuron Growth under Electrical Stimulation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4794982. [PMID: 32337253 PMCID: PMC7153003 DOI: 10.1155/2020/4794982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 12/20/2022]
Abstract
Objectives To prepare the conductive MWCNT (multiwall carbon nanotube)-agarose scaffolds with multi-microchannel for neuron growth under electrical stimulation. Methods The scaffolds were produced by gradient freeze and lyophilization methods. The synthesized materials were characterized by SEM and near-infrared spectroscopy, and their microstructure, swelling-deswelling, conductivity, biocompatibility, and shape memory behavior were measured. A three-dimensional culture model by implanting cells into scaffolds was built, and the behaviors of RSC96 cells on scaffolds under electrical stimulation were evaluated. Results The addition of MWCNT did not affect the pore composition ratio and shape memory of agarose scaffolds, but 0.025% wt MWCNT in scaffolds improved the swelling ratio and water retention at the swelling equilibrium state. Though MWCNTs in high concentration had slight effect on proliferation of RSC96 cells and PC12 cells, there was no difference that the expressions of neurofilament of RSC96 cells on scaffolds with MWCNTs of different concentration. RSC96 cells arranged better along the longitudinal axis of scaffolds and showed better adhesion on both 0.025% MWCNT-agarose scaffolds and 0.05% MWCNT-agarose scaffolds compared to other scaffolds. Conclusions Agarose scaffolds with MWCNTs possessed promising applicable prospect in peripheral nerve defects.
Collapse
|
29
|
Qi D, Shi W, Black AR, Kuss MA, Pang X, He Y, Liu B, Duan B. Repair and regeneration of small intestine: A review of current engineering approaches. Biomaterials 2020; 240:119832. [PMID: 32113114 DOI: 10.1016/j.biomaterials.2020.119832] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 02/06/2023]
Abstract
The small intestine (SI) is difficult to regenerate or reconstruct due to its complex structure and functions. Recent developments in stem cell research, advanced engineering technologies, and regenerative medicine strategies bring new hope of solving clinical problems of the SI. This review will first summarize the structure, function, development, cell types, and matrix components of the SI. Then, the major cell sources for SI regeneration are introduced, and state-of-the-art biofabrication technologies for generating engineered SI tissues or models are overviewed. Furthermore, in vitro models and in vivo transplantation, based on intestinal organoids and tissue engineering, are highlighted. Finally, current challenges and future perspectives are discussed to help direct future applications for SI repair and regeneration.
Collapse
Affiliation(s)
- Dianjun Qi
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell A Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xining Pang
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China; Department of Academician Expert Workstation and Liaoning Province Human Amniotic Membrane Dressings Stem Cells and Regenerative Medicine Engineering Research Center, Shenyang Amnion Biological Engineering Technology Research and Development Center Co., Ltd, Shenyang, Liaoning, China
| | - Yini He
- Department of General Practice, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bing Liu
- Department of Anorectal Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
30
|
Ferracci G, Zhu M, Ibrahim MS, Ma G, Fan TF, Lee BH, Cho NJ. Photocurable Albumin Methacryloyl Hydrogels as a Versatile Platform for Tissue Engineering. ACS APPLIED BIO MATERIALS 2020; 3:920-934. [DOI: 10.1021/acsabm.9b00984] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Gaia Ferracci
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Mengxiang Zhu
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Wenzhou Institute of Biomaterials and Engineering, University of CAS, Wenzhou, Zhejiang 325011, China
| | - Mohammed Shahrudin Ibrahim
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Gamaliel Ma
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Teng Fei Fan
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Bae Hoon Lee
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Wenzhou Institute of Biomaterials and Engineering, University of CAS, Wenzhou, Zhejiang 325011, China
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| |
Collapse
|
31
|
Pavlova E, Nikishin I, Bogdanova A, Klinov D, Bagrov D. The miscibility and spatial distribution of the components in electrospun polymer–protein mats. RSC Adv 2020; 10:4672-4680. [PMID: 35495279 PMCID: PMC9049090 DOI: 10.1039/c9ra10910b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 01/17/2020] [Indexed: 01/02/2023] Open
Abstract
Biodegradable blended electrospun mats are promising for biomedical applications such as wound dressing, tissue engineering, and drug delivery. Electrospun mats based on polyesters can be modified by the addition of other polymers or proteins to accelerate the degradation, improve mechanical properties or biocompatibility. However, relatively little is known about the distribution of the components throughout the blended mats. In the present work, we prepared polylactide (PLA), bovine serum albumin (BSA), and the blended PLA–BSA electrospun mats. We demonstrated that PLA and BSA are miscible in a common solvent HFIP (1,1,1,3,3,3-hexafluoro-2-propanol) at concentrations below 3%, but become immiscible as concentration increases. We used three methods (fluorescence microscopy, EDX, and Raman microspectroscopy) to validate that PLA and BSA can be blended in a single electrospun fiber despite the phase separation in the blend. The homogeneity of the blend influences on the homogeneity of the distribution of PLA and BSA components throughout the electrospun mat, as measured by Raman microspectroscopy. When the blended electrospun mats were incubated in water, they demonstrated the prolonged release of BSA. The presented results show a step-by-step approach for manufacturing blended electrospun mats made of immiscible components, which involves the analysis of component miscibility, the mat morphology, and composition. This approach can be used for the rational design of multicomponent electrospun mats. Polylactide and bovine serum albumin can be combined in a single electrospun fiber, despite the phase separation.![]()
Collapse
Affiliation(s)
- Elizaveta Pavlova
- Federal Research Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency of Russia
- Moscow
- Russian Federation
- Moscow Institute of Physics and Technology
- Moscow Region
| | - Igor Nikishin
- Lomonosov Moscow State University
- Faculty of Biology
- Department of Bioengineering
- Moscow
- Russian Federation
| | - Alexandra Bogdanova
- Federal Research Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency of Russia
- Moscow
- Russian Federation
- Moscow Institute of Physics and Technology
- Moscow Region
| | - Dmitry Klinov
- Federal Research Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency of Russia
- Moscow
- Russian Federation
- Moscow Institute of Physics and Technology
- Moscow Region
| | - Dmitry Bagrov
- Federal Research Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency of Russia
- Moscow
- Russian Federation
- Lomonosov Moscow State University
- Faculty of Biology
| |
Collapse
|
32
|
Yan H, Wang Y, Li L, Zhou X, Shi X, Wei Y, Zhang P. A micropatterned conductive electrospun nanofiber mesh combined with electrical stimulation for synergistically enhancing differentiation of rat neural stem cells. J Mater Chem B 2020; 8:2673-2688. [DOI: 10.1039/c9tb02864a] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The micropatterned conductive nanofiber mesh combined with ES effectively facilitates the differentiation of NSCs into neuron and suppresses the formation of astrocytes.
Collapse
Affiliation(s)
- Huanhuan Yan
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Yu Wang
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Linlong Li
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Xiaosong Zhou
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Xincui Shi
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| | - Yen Wei
- Department of Chemistry
- Tsinghua University
- Beijing 100084
- P. R. China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- P. R. China
| |
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
|
Sharma P, Kaur H, Roy S. Designing a Tenascin-C-Inspired Short Bioactive Peptide Scaffold to Direct and Control Cellular Behavior. ACS Biomater Sci Eng 2019; 5:6497-6510. [DOI: 10.1021/acsbiomaterials.9b01115] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Pooja Sharma
- Institute of Nanoscience and Technology, Habitat Centre, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Harsimran Kaur
- Institute of Nanoscience and Technology, Habitat Centre, Sector 64, Phase 10, Mohali, Punjab 160062, India
| | - Sangita Roy
- Institute of Nanoscience and Technology, Habitat Centre, Sector 64, Phase 10, Mohali, Punjab 160062, India
| |
Collapse
|
35
|
Ong J, Zhao J, Justin AW, Markaki AE. Albumin-based hydrogels for regenerative engineering and cell transplantation. Biotechnol Bioeng 2019; 116:3457-3468. [PMID: 31520415 PMCID: PMC6899591 DOI: 10.1002/bit.27167] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 01/04/2023]
Abstract
Albumin, the most abundant plasma protein in mammals, is a versatile and easily obtainable biomaterial. It is pH and temperature responsive, dissolvable in high concentrations and gels readily in defined conditions. This versatility, together with its inexpensiveness and biocompatibility, makes albumin an attractive biomaterial for biomedical research and therapeutics. So far, clinical research in albumin has centered mainly on its use as a carrier molecule or nanoparticle to improve drug pharmacokinetics and delivery to target sites. In contrast, research in albumin-based hydrogels is less established albeit growing in interest over recent years. In this minireview, we report current literature and critically discuss the synthesis, mechanical properties, biological effects and uses, biodegradability and cost of albumin hydrogels as a xeno-free, customizable, and transplantable construct for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- John Ong
- Department of Engineering, University of Cambridge, Cambridge, UK.,Gastroenterology Specialty Training Program, East of England Deanery, Cambridge, UK
| | - Junzhe Zhao
- Department of Engineering, University of Cambridge, Cambridge, UK
| | | | - Athina E Markaki
- Department of Engineering, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Slepchenkov MM, Gerasimenko AY, Telyshev DV, Glukhova OE. Protein-Polymer Matrices with Embedded Carbon Nanotubes for Tissue Engineering: Regularities of Formation and Features of Interaction with Cell Membranes. MATERIALS 2019; 12:ma12193083. [PMID: 31546631 PMCID: PMC6803951 DOI: 10.3390/ma12193083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/12/2019] [Accepted: 09/17/2019] [Indexed: 12/17/2022]
Abstract
This paper reveals the mechanism of nanowelding a branched network of single-walled carbon nanotubes (SWCNTs) used as a framework for the formation of protein-polymer matrices with albumin, collagen, and chitosan. It is shown that the introduction of certain point defects into the structure of SWCNTs (single vacancy, double vacancy, Stone-Wales defect, and a mixed defect) allows us to obtain strong heating in defective regions as compared to ideal SWCNTs. The wavelengths at which absorption reaches 50% are determined. Non-uniform absorption of laser radiation along with inefficient heat removal in defective regions determines the formation of hot spots, in which nanowelding of SWCNTs is observed even at 0.36 nm between contacting surfaces. The regularities of formation of layered protein-polymer matrices and the features of their interaction with cell membrane are revealed. All studies are carried out in silico using high-precision quantum approaches.
Collapse
Affiliation(s)
- Michael M Slepchenkov
- Department of Physics, Saratov State University, Astrakhanskaya street 83, Saratov 410012, Russia.
| | - Alexander Yu Gerasimenko
- Laboratory of Biomedical Nanotechnology, I.M. Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya street 2-4, Moscow 119991, Russia.
- Institute of Biomedical Systems, National Research University of Electronic Technology MIET, Shokin Square 1, Zelenograd, Moscow 124498, Russia.
| | - Dmitry V Telyshev
- Laboratory of Biomedical Nanotechnology, I.M. Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya street 2-4, Moscow 119991, Russia.
- Institute of Biomedical Systems, National Research University of Electronic Technology MIET, Shokin Square 1, Zelenograd, Moscow 124498, Russia.
| | - Olga E Glukhova
- Department of Physics, Saratov State University, Astrakhanskaya street 83, Saratov 410012, Russia.
- Laboratory of Biomedical Nanotechnology, I.M. Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya street 2-4, Moscow 119991, Russia.
| |
Collapse
|
37
|
Zhang D, Wang Y. Functional Protein-Based Bioinspired Nanomaterials: From Coupled Proteins, Synthetic Approaches, Nanostructures to Applications. Int J Mol Sci 2019; 20:E3054. [PMID: 31234528 PMCID: PMC6627797 DOI: 10.3390/ijms20123054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 11/16/2022] Open
Abstract
Protein-based bioinspired nanomaterials (PBNs) combines the advantage of the size, shape, and surface chemistry of nanomaterials, the morphology and functions of natural materials, and the physical and chemical properties of various proteins. Recently, there are many exciting developments on biomimetic nanomaterials using proteins for different applications including, tissue engineering, drug delivery, diagnosis and therapy, smart materials and structures, and water collection and separation. Protein-based biomaterials with high biocompatibility and biodegradability could be modified to obtain the healing effects of natural organisms after injury by mimicking the extracellular matrix. For cancer and other diseases that are difficult to cure now, new therapeutic methods involving different kinds of biomaterials are studied. The nanomaterials with surface modification, which can achieve high drug loading, can be used as drug carriers to enhance target and trigger deliveries. For environment protection and the sustainability of the world, protein-based nanomaterials are also applied for water treatment. A wide range of contaminants from natural water source, such as organic dyes, oil substances, and multiple heavy ions, could be absorbed by protein-based nanomaterials. This review summarizes the formation and application of functional PBNs, and the details of their nanostructures, the proteins involved, and the synthetic approaches are addressed.
Collapse
Affiliation(s)
- Dong Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Hum, Kowloon 999077, Hong Kong.
| | - Yi Wang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Hum, Kowloon 999077, Hong Kong.
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation) and Shenzhen Key Laboratory of Food Biological Safety Control, Shenzhen Research Institute of Hong Kong Polytechnic University, Shenzhen 518057, China.
| |
Collapse
|
38
|
Abstract
Electrospinning is a versatile and viable technique for generating ultrathin fibers. Remarkable progress has been made with regard to the development of electrospinning methods and engineering of electrospun nanofibers to suit or enable various applications. We aim to provide a comprehensive overview of electrospinning, including the principle, methods, materials, and applications. We begin with a brief introduction to the early history of electrospinning, followed by discussion of its principle and typical apparatus. We then discuss its renaissance over the past two decades as a powerful technology for the production of nanofibers with diversified compositions, structures, and properties. Afterward, we discuss the applications of electrospun nanofibers, including their use as "smart" mats, filtration membranes, catalytic supports, energy harvesting/conversion/storage components, and photonic and electronic devices, as well as biomedical scaffolds. We highlight the most relevant and recent advances related to the applications of electrospun nanofibers by focusing on the most representative examples. We also offer perspectives on the challenges, opportunities, and new directions for future development. At the end, we discuss approaches to the scale-up production of electrospun nanofibers and briefly discuss various types of commercial products based on electrospun nanofibers that have found widespread use in our everyday life.
Collapse
Affiliation(s)
- Jiajia Xue
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Tong Wu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | - Yunqian Dai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, People’s Republic of China
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- School of Chemistry and Biochemistry, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
39
|
Ye K, Kuang H, You Z, Morsi Y, Mo X. Electrospun Nanofibers for Tissue Engineering with Drug Loading and Release. Pharmaceutics 2019; 11:E182. [PMID: 30991742 PMCID: PMC6523318 DOI: 10.3390/pharmaceutics11040182] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/03/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
Electrospinning technologies have been applied in the field of tissue engineering as materials, with nanoscale-structures and high porosity, can be easily prepared via this method to bio-mimic the natural extracellular matrix (ECM). Tissue engineering aims to fabricate functional biomaterials for the repairment and regeneration of defective tissue. In addition to the structural simulation for accelerating the repair process and achieving a high-quality regeneration, the combination of biomaterials and bioactive molecules is required for an ideal tissue-engineering scaffold. Due to the diversity in materials and method selection for electrospinning, a great flexibility in drug delivery systems can be achieved. Various drugs including antibiotic agents, vitamins, peptides, and proteins can be incorporated into electrospun scaffolds using different electrospinning techniques and drug-loading methods. This is a review of recent research on electrospun nanofibrous scaffolds for tissue-engineering applications, the development of preparation methods, and the delivery of various bioactive molecules. These studies are based on the fabrication of electrospun biomaterials for the repair of blood vessels, nerve tissues, cartilage, bone defects, and the treatment of aneurysms and skin wounds, as well as their applications related to oral mucosa and dental fields. In these studies, due to the optimal selection of drugs and loading methods based on electrospinning, in vitro and in vivo experiments demonstrated that these scaffolds exhibited desirable effects for the repair and treatment of damaged tissue and, thus, have excellent potential for clinical application.
Collapse
Affiliation(s)
- Kaiqiang Ye
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Haizhu Kuang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Yosry Morsi
- Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, Boroondara, VIC 3122, Australia.
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| |
Collapse
|
40
|
Mobini S, Song YH, McCrary MW, Schmidt CE. Advances in ex vivo models and lab-on-a-chip devices for neural tissue engineering. Biomaterials 2019; 198:146-166. [PMID: 29880219 PMCID: PMC6957334 DOI: 10.1016/j.biomaterials.2018.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/25/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023]
Abstract
The technologies related to ex vivo models and lab-on-a-chip devices for studying the regeneration of brain, spinal cord, and peripheral nerve tissues are essential tools for neural tissue engineering and regenerative medicine research. The need for ex vivo systems, lab-on-a-chip technologies and disease models for neural tissue engineering applications are emerging to overcome the shortages and drawbacks of traditional in vitro systems and animal models. Ex vivo models have evolved from traditional 2D cell culture models to 3D tissue-engineered scaffold systems, bioreactors, and recently organoid test beds. In addition to ex vivo model systems, we discuss lab-on-a-chip devices and technologies specifically for neural tissue engineering applications. Finally, we review current commercial products that mimic diseased and normal neural tissues, and discuss the future directions in this field.
Collapse
Affiliation(s)
- Sahba Mobini
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Michaela W McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
41
|
Shi X, Wu H, Yan H, Wang Y, Wang Z, Zhang P. Electroactive Nanocomposite Porous Scaffolds of PAPn/op-HA/PLGA Enhance Osteogenesis in Vivo. ACS APPLIED BIO MATERIALS 2019; 2:1464-1476. [DOI: 10.1021/acsabm.8b00716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xincui Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Haitao Wu
- Department of Orthopedics, Jilin Provincial People’s Hospital, 1183 Gongnong Street, Changchun 130021, China
| | - Huanhuan Yan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Yu Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
42
|
Amdursky N, Głowacki ED, Meredith P. Macroscale Biomolecular Electronics and Ionics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1802221. [PMID: 30334284 DOI: 10.1002/adma.201802221] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/25/2018] [Indexed: 05/18/2023]
Abstract
The conduction of ions and electrons over multiple length scales is central to the processes that drive the biological world. The multidisciplinary attempts to elucidate the physics and chemistry of electron, proton, and ion transfer in biological charge transfer have focused primarily on the nano- and microscales. However, recently significant progress has been made on biomolecular materials that can support ion and electron currents over millimeters if not centimeters. Likewise, similar transport phenomena in organic semiconductors and ionics have led to new innovations in a wide variety of applications from energy generation and storage to displays and bioelectronics. Here, the underlying principles of conduction on the macroscale in biomolecular materials are discussed, highlighting recent examples, and particularly the establishment of accurate structure-property relationships to guide rationale material and device design. The technological viability of biomolecular electronics and ionics is also discussed.
Collapse
Affiliation(s)
- Nadav Amdursky
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Eric Daniel Głowacki
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Bredgatan 33, SE-60174, Norrköping, Sweden
- Wallenberg Centre for Molecular Medicine, Linköping University, 58183, Linköping, Sweden
| | - Paul Meredith
- Department of Physics, Swansea University, Singleton Park, Swansea, SA2 8PP, Wales, UK
| |
Collapse
|
43
|
Amdursky N, Mazo MM, Thomas MR, Humphrey EJ, Puetzer JL, St-Pierre JP, Skaalure SC, Richardson RM, Terracciano CM, Stevens MM. Elastic serum-albumin based hydrogels: mechanism of formation and application in cardiac tissue engineering. J Mater Chem B 2018; 6:5604-5612. [PMID: 30283632 PMCID: PMC6166857 DOI: 10.1039/c8tb01014e] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hydrogels are promising materials for mimicking the extra-cellular environment. Here, we present a simple methodology for the formation of a free-standing viscoelastic hydrogel from the abundant and low cost protein serum albumin. We show that the mechanical properties of the hydrogel exhibit a complicated behaviour as a function of the weight fraction of the protein component. We further use X-ray scattering to shed light on the mechanism of gelation from the formation of a fibrillary network at low weight fractions to interconnected aggregates at higher weight fractions. Given the match between our hydrogel elasticity and that of the myocardium, we investigated its potential for supporting cardiac cells in vitro. Interestingly, these hydrogels support the formation of several layers of myocytes and significantly promote the maintenance of a native-like gene expression profile compared to those cultured on glass. When confronted with a multicellular ventricular cell preparation, the hydrogels can support macroscopically contracting cardiac-like tissues with a distinct cell arrangement, and form mm-long vascular-like structures. We envisage that our simple approach for the formation of an elastic substrate from an abundant protein makes the hydrogel a compelling biomedical material candidate for a wide range of cell types.
Collapse
Affiliation(s)
- Nadav Amdursky
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| | - Manuel M. Mazo
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| | - Michael R. Thomas
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| | - Eleanor J. Humphrey
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Jennifer L. Puetzer
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| | - Jean-Philippe St-Pierre
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| | - Stacey C. Skaalure
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| | | | - Cesare M. Terracciano
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College Lodon, London, SW7 2AZ, UK
| |
Collapse
|