1
|
Liu Z, Jia J, Lei Q, Wei Y, Hu Y, Lian X, Zhao L, Xie X, Bai H, He X, Si L, Livermore C, Kuang R, Zhang Y, Wang J, Yu Z, Ma X, Huang D. Electrohydrodynamic Direct-Writing Micro/Nanofibrous Architectures: Principle, Materials, and Biomedical Applications. Adv Healthc Mater 2024; 13:e2400930. [PMID: 38847291 DOI: 10.1002/adhm.202400930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/21/2024] [Indexed: 07/05/2024]
Abstract
Electrohydrodynamic (EHD) direct-writing has recently gained attention as a highly promising additive manufacturing strategy for fabricating intricate micro/nanoscale architectures. This technique is particularly well-suited for mimicking the extracellular matrix (ECM) present in biological tissue, which serves a vital function in facilitating cell colonization, migration, and growth. The integration of EHD direct-writing with other techniques has been employed to enhance the biological performance of scaffolds, and significant advancements have been made in the development of tailored scaffold architectures and constituents to meet the specific requirements of various biomedical applications. Here, a comprehensive overview of EHD direct-writing is provided, including its underlying principles, demonstrated materials systems, and biomedical applications. A brief chronology of EHD direct-writing is provided, along with an examination of the observed phenomena that occur during the printing process. The impact of biomaterial selection and architectural topographic cues on biological performance is also highlighted. Finally, the major limitations associated with EHD direct-writing are discussed.
Collapse
Affiliation(s)
- Zhengjiang Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Jinqiao Jia
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
| | - Qi Lei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Yinchun Hu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| | - Xin Xie
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Haiqing Bai
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Xiaomin He
- Xellar Biosystems, Cambridge, MA, 02458, USA
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Carol Livermore
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Rong Kuang
- Zhejiang Institute for Food and Drug Control, Hangzhou, 310000, P. R. China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Jiucun Wang
- Human Phenome Institute, Fudan University, Shanghai, 200433, P. R. China
| | - Zhaoyan Yu
- Shandong Public Health Clinical Center, Shandong University, Jinan, 250000, P. R. China
| | - Xudong Ma
- Cytori Therapeutics LLC., Shanghai, 201802, P. R. China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of biomedical Engineering, Taiyuan University of Technology, Taiyuan, 030024, P. R. China
- Shanxi-Zheda Institute of advanced Materials and Chemical Engineering, Taiyuan, 030032, P. R. China
| |
Collapse
|
2
|
Akgul B, Gulcan C, Tornaci S, Erginer M, Toksoy Oner E, Abamor ES, Acar S, Allahverdiyev AM. Manufacturing Radially Aligned PCL Nanofibers Reinforced With Sulfated Levan and Evaluation of its Biological Activity for Healing Tympanic Membrane Perforations. Macromol Biosci 2024:e2400291. [PMID: 39461894 DOI: 10.1002/mabi.202400291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/08/2024] [Indexed: 10/29/2024]
Abstract
The main objective of this study is to construct radially aligned PCL nanofibers reinforced with levan polymer and investigate their in vitro biological activities thoroughly. First Halomonas levan (HL) polysaccharide is hydrolyzed (hHL) and subjected to sulfation to attain Sulfated hydrolyzed Halomonas levan (ShHL)-based material indicating heparin mimetic properties. Then, optimization studies are carried out to produce coaxially generated radially aligned Poly(caprolactone) (PCL) -ShHL nanofibers via electrospinning. The obtained nanofibers are characterized with Fourier Transform Infrared Spectroscopy (FTIR) and Field Emission Scanning Electron Microscopy with Energy Dispersive X-Ray (FESEM-EDX) analysis, and mechanical, contact angle measurement, biodegradability, and swelling tests as well. Afterward, cytotoxicity of artificial tympanic membranes is analyzed by MTT (3-(4,5-Dimethylthiazol-2-yl) -2,5 Diphenyltetrazolium Bromide) test, and their impacts on cell proliferation, cellular adhesion, wound healing processes are explored. Furthermore, an additional FESEM imaging is performed to manifest the interactions between fibroblasts and nanofibers. According to analytical measurements it is detected that PCL-ShHL nanofibers i) are smaller in fiber diameter, ii) are more biodegradable, iii) are more hydrophilic, and iv) demonstrated superior mechanical properties compared to PCL nanofibers. Moreover, it is also deciphered that PCL-ShHL nanofibers strongly elevated cellular adhesion, proliferation, and in vitro wound healing features compared to PCL nanofibers. According to obtained results it is assumed that newly synthetized levan and PCL mediated nanofibers are very encouraging for healing tympanic membrane perforations.
Collapse
Affiliation(s)
- Busra Akgul
- Department of Bioengineering, Yildiz Technical University, Istanbul, 34220, Turkey
| | - Cansu Gulcan
- Department of Bioengineering, Yildiz Technical University, Istanbul, 34220, Turkey
| | - Selay Tornaci
- IBSB, Department of Bioengineering, Marmara University, Istanbul, 34854, Turkey
| | - Merve Erginer
- Institute of Nanotechnology and Biotechnology, Istanbul University-Cerrahpaşa, Istanbul, 34500, Turkey
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, Istanbul, 34220, Turkey
| | - Ebru Toksoy Oner
- IBSB, Department of Bioengineering, Marmara University, Istanbul, 34854, Turkey
| | - Emrah Sefik Abamor
- Department of Bioengineering, Yildiz Technical University, Istanbul, 34220, Turkey
| | - Serap Acar
- Department of Bioengineering, Yildiz Technical University, Istanbul, 34220, Turkey
| | - Adil M Allahverdiyev
- The V. Akhundov Scientific Research Medical Preventive Institute, Baku, AZ1004, Azerbaijan
| |
Collapse
|
3
|
Fatima Balderrama I, Schafer S, El Shatanofy M, Bergamo ETP, Mirsky NA, Nayak VV, Marcantonio Junior E, Alifarag AM, Coelho PG, Witek L. Biomimetic Tissue Engineering Strategies for Craniofacial Applications. Biomimetics (Basel) 2024; 9:636. [PMID: 39451842 PMCID: PMC11506466 DOI: 10.3390/biomimetics9100636] [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: 08/15/2024] [Revised: 09/21/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Biomimetics is the science of imitating nature's designs and processes to create innovative solutions for various fields, including dentistry and craniofacial reconstruction. In these areas, biomimetics involves drawing inspiration from living organisms/systems to develop new materials, techniques, and devices that closely resemble natural tissue structures and enhance functionality. This field has successfully demonstrated its potential to revolutionize craniofacial procedures, significantly improving patient outcomes. In dentistry, biomimetics offers exciting possibilities for the advancement of new dental materials, restorative techniques, and regenerative potential. By analyzing the structure/composition of natural teeth and the surrounding tissues, researchers have developed restorative materials that mimic the properties of teeth, as well as regenerative techniques that might assist in repairing enamel, dentin, pulp, cementum, periodontal ligament, and bone. In craniofacial reconstruction, biomimetics plays a vital role in developing innovative solutions for facial trauma, congenital defects, and various conditions affecting the maxillofacial region. By studying the intricate composition and mechanical properties of the skull and facial bones, clinicians and engineers have been able to replicate natural structures leveraging computer-aided design and manufacturing (CAD/CAM) and 3D printing. This has allowed for the creation of patient-specific scaffolds, implants, and prostheses that accurately fit a patient's anatomy. This review highlights the current evidence on the application of biomimetics in the fields of dentistry and craniofacial reconstruction.
Collapse
Affiliation(s)
- Isis Fatima Balderrama
- Department of Diagnosis and Surgery, School of Dentistry of Araraquara, Sao Paulo State University, Sao Paulo 14801-385, Brazil
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
| | - Sogand Schafer
- Division of Plastic, Reconstructive and Oral Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Muhammad El Shatanofy
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Edmara T. P. Bergamo
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Prosthodontics, NYU Dentistry, New York, NY 10010, USA
| | | | - Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Elcio Marcantonio Junior
- Department of Diagnosis and Surgery, School of Dentistry of Araraquara, Sao Paulo State University, Sao Paulo 14801-385, Brazil
| | - Adham M. Alifarag
- Department of General Surgery, Temple University Hospital System, Philadelphia, PA 19140, USA
| | - Paulo G. Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Plastic Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, USA
- Hansjörg Wyss Department of Plastic Surgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
4
|
Lu M, Xu Z, Xu F, Yin C, Guo H, Cheng B. Mechanical network motifs as targets for mechanomedicine. Drug Discov Today 2024; 29:104145. [PMID: 39182599 DOI: 10.1016/j.drudis.2024.104145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The identification and analysis of network motifs has been widely used in the functional analysis of signaling components, disease discovery and other fields. The positive feedback loop (PFL) is a simple but important network motif. The formation of a PFL is regulated by mechanical cues such as substrate stiffness, fiber stretching and cell compression in the cell microenvironment. Here, we propose a new term, 'mechanical PFL', and analyze the mechanisms of mechanical PFLs at molecular, subcellular and cellular scales. More and more therapies are being targeted against mechanosignaling pathways at the experimental and preclinical stages, and exploring mechanical PFLs as potential mechanomedicine targets could be a new direction for disease treatment.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zhao Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China.
| | - Hui Guo
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China.
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
5
|
Zhang Y, Chen J, Sun Y, Wang M, Liu H, Zhang W. Endogenous Tissue Engineering for Chondral and Osteochondral Regeneration: Strategies and Mechanisms. ACS Biomater Sci Eng 2024; 10:4716-4739. [PMID: 39091217 DOI: 10.1021/acsbiomaterials.4c00603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Increasing attention has been paid to the development of effective strategies for articular cartilage (AC) and osteochondral (OC) regeneration due to their limited self-reparative capacities and the shortage of timely and appropriate clinical treatments. Traditional cell-dependent tissue engineering faces various challenges such as restricted cell sources, phenotypic alterations, and immune rejection. In contrast, endogenous tissue engineering represents a promising alternative, leveraging acellular biomaterials to guide endogenous cells to the injury site and stimulate their intrinsic regenerative potential. This review provides a comprehensive overview of recent advancements in endogenous tissue engineering strategies for AC and OC regeneration, with a focus on the tissue engineering triad comprising endogenous stem/progenitor cells (ESPCs), scaffolds, and biomolecules. Multiple types of ESPCs present within the AC and OC microenvironment, including bone marrow-derived mesenchymal stem cells (BMSCs), adipose-derived mesenchymal stem cells (AD-MSCs), synovial membrane-derived mesenchymal stem cells (SM-MSCs), and AC-derived stem/progenitor cells (CSPCs), exhibit the ability to migrate toward injury sites and demonstrate pro-regenerative properties. The fabrication and characteristics of scaffolds in various formats including hydrogels, porous sponges, electrospun fibers, particles, films, multilayer scaffolds, bioceramics, and bioglass, highlighting their suitability for AC and OC repair, are systemically summarized. Furthermore, the review emphasizes the pivotal role of biomolecules in facilitating ESPCs migration, adhesion, chondrogenesis, osteogenesis, as well as regulating inflammation, aging, and hypertrophy-critical processes for endogenous AC and OC regeneration. Insights into the applications of endogenous tissue engineering strategies for in vivo AC and OC regeneration are provided along with a discussion on future perspectives to enhance regenerative outcomes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009 Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009 Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096 Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), 310058 Hangzhou, China
| |
Collapse
|
6
|
Farahani PK. Application of Tissue Engineering and Biomaterials in Nose Surgery. JPRAS Open 2024; 40:262-272. [PMID: 38708386 PMCID: PMC11067003 DOI: 10.1016/j.jpra.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 05/07/2024] Open
Abstract
Surgery of the nose involves a series of operations that are directed at restoring the nasal anatomy and physiology. The extent or degree of reconstruction needed is dependent on the appearance-based requirement of the patients and the procedure exploited for the correction such that nasal airflow is preserved. Standard surgical approach includes the use of autologous tissue or implantation alloplastic bio or synthetic/fabricated construct materials to correct the defects. Over the years, tissue engineering has been proven to be a promising technique for reconstructing tissue and organ defects, including the nose. Recently, there has been keen interest in fabricating new tissues and organ scaffolds using 3D printing technology with good control over the micro-architecture and excellent interior architecture suitable for cell seeding. Unviability of the tissue and harvest-associated complications have increased the need for the investigation of tissue engineering based methods for nasal reconstruction using biomaterials, stem cells, and growth factors combined with 3D bioprinting. However, there are only a handful of studies vis-à-vis the application of cartilage tissue engineering, stem cells, and growth factors for the purpose. This review provides highlights about the available studies based on the application of stem cells, biomaterials, and growth factors for nasal reconstruction surgery, as there is limited recent information on the use of these entities in nasal surgeries.
Collapse
|
7
|
Koshy J, Sangeetha D. Recent progress and treatment strategy of pectin polysaccharide based tissue engineering scaffolds in cancer therapy, wound healing and cartilage regeneration. Int J Biol Macromol 2024; 257:128594. [PMID: 38056744 DOI: 10.1016/j.ijbiomac.2023.128594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/12/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
Natural polymers and its mixtures in the form of films, sponges and hydrogels are playing a major role in tissue engineering and regenerative medicine. Hydrogels have been extensively investigated as standalone materials for drug delivery purposes as they enable effective encapsulation and sustained release of drugs. Biopolymers are widely utilised in the fabrication of hydrogels due to their safety, biocompatibility, low toxicity, and regulated breakdown by human enzymes. Among all the biopolymers, polysaccharide-based polymer is well suited to overcome the limitations of traditional wound dressing materials. Pectin is a polysaccharide which can be extracted from different plant sources and is used in various pharmaceutical and biomedical applications including cartilage regeneration. Pectin itself cannot be employed as scaffolds for tissue engineering since it decomposes quickly. This article discusses recent research and developments on pectin polysaccharide, including its types, origins, applications, and potential demands for use in AI-mediated scaffolds. It also covers the materials-design process, strategy for implementation to material selection and fabrication methods for evaluation. Finally, we discuss unmet requirements and current obstacles in the development of optimal materials for wound healing and bone-tissue regeneration, as well as emerging strategies in the field.
Collapse
Affiliation(s)
- Jijo Koshy
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - D Sangeetha
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
8
|
Asadian M, Tomasina C, Onyshchenko Y, Chan KV, Norouzi M, Zonderland J, Camarero-Espinosa S, Morent R, De Geyter N, Moroni L. The role of plasma-induced surface chemistry on polycaprolactone nanofibers to direct chondrogenic differentiation of human mesenchymal stem cells. J Biomed Mater Res A 2024; 112:210-230. [PMID: 37706337 DOI: 10.1002/jbm.a.37607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023]
Abstract
Bone marrow-derived mesenchymal stromal cells (BMSCs) are extensively being utilized for cartilage regeneration owing to their excellent differentiation potential and availability. However, controlled differentiation of BMSCs towards cartilaginous phenotypes to heal full-thickness cartilage defects remains challenging. This study investigates how different surface properties induced by either coating deposition or biomolecules immobilization onto nanofibers (NFs) could affect BMSCs chondro-inductive behavior. Accordingly, electrospun poly(ε-caprolactone) (PCL) NFs were exposed to two surface modification strategies based on medium-pressure plasma technology. The first strategy is plasma polymerization, in which cyclopropylamine (CPA) or acrylic acid (AcAc) monomers were plasma polymerized to obtain amine- or carboxylic acid-rich NFs, respectively. The second strategy uses a combination of CPA plasma polymerization and a post-chemical technique to immobilize chondroitin sulfate (CS) onto the NFs. These modifications could affect surface roughness, hydrophilicity, and chemical composition while preserving the NFs' nano-morphology. The results of long-term BMSCs culture in both basic and chondrogenic media proved that the surface modifications modulated BMSCs chondrogenic differentiation. Indeed, the incorporation of polar groups by different modification strategies had a positive impact on the cell proliferation rate, production of the glycosaminoglycan matrix, and expression of extracellular matrix proteins (collagen I and collagen II). The chondro-inductive behavior of the samples was highly dependent on the nature of the introduced polar functional groups. Among all samples, carboxylic acid-rich NFs promoted chondrogenesis by higher expression of aggrecan, Sox9, and collagen II with downregulation of hypertrophic markers. Hence, this approach showed an intrinsic potential to have a non-hypertrophic chondrogenic cell phenotype.
Collapse
Affiliation(s)
- Mahtab Asadian
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
- Prometheus Division of Skeletal Tissue Engineering, Department of Materials Science, KU Leuven University, Leuven, Belgium
| | - Clarissa Tomasina
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| | - Yuliia Onyshchenko
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Ke Vin Chan
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Mohammad Norouzi
- Department of Pharmacology, University of Montreal, Montreal, Québec, Canada
| | - Jip Zonderland
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| | - Sandra Camarero-Espinosa
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
- POLYMAT University of the Basque Country UPV/EHU Avenida Tolosa 72, Donostia/San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, Euskadi Pl. 5, Bilbao, Spain
| | - Rino Morent
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Nathalie De Geyter
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Ghent University, Ghent, Belgium
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration Department, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
9
|
Howard CJ, Paul A, Duruanyanwu J, Sackho K, Campagnolo P, Stolojan V. The Manufacturing Conditions for the Direct and Reproducible Formation of Electrospun PCL/Gelatine 3D Structures for Tissue Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:3107. [PMID: 38133004 PMCID: PMC10745430 DOI: 10.3390/nano13243107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
Electrospinning is a versatile technique for fabricating nanofibrous scaffolds for tissue engineering applications. However, the direct formation of 3D sponges through electrospinning has previously not been reproducible. We used a Taguchi experimental design approach to optimise the electrospinning parameters for forming PCL and PCL/gelatine 3D sponges. The following parameters were investigated to improve sponge formation: solution concentration, humidity, and solution conductivity. Pure PCL sponges were achievable. However, a much fluffier sponge formed by increasing the solution conductivity with gelatine. The optimal conditions for sponge formation 24 w/v% 80:20 PCL:gelatine on aluminium foil at ≥70% humidity, 15 cm, 22 kV and 1500 µL/h. The resulting sponge had a highly porous structure with a fibre diameter of ~1 µm. They also supported significantly higher cell viability than 2D electrospun mats, dropcast films of the same material and even the TCP positive control. Our study demonstrates that the direct formation of PCL/gelatine 3D sponges through electrospinning is feasible and promising for tissue engineering applications. The sponges have a highly porous structure and support cell viability, which are essential properties for tissue engineering scaffolds. Further studies are needed to optimise the manufacturing process and evaluate the sponges' long-term performance in vivo.
Collapse
Affiliation(s)
- Chloe Jayne Howard
- Advanced Technology Institute, School of Computer Science and Electronic Engineering, University of Surrey, Guildford GU2 7XH, UK; (C.J.H.); (A.P.)
| | - Aumrita Paul
- Advanced Technology Institute, School of Computer Science and Electronic Engineering, University of Surrey, Guildford GU2 7XH, UK; (C.J.H.); (A.P.)
| | - Justin Duruanyanwu
- Department of Biochemical Sciences, University of Surrey, Guildford GU2 7XH, UK; (J.D.); (K.S.); (P.C.)
| | - Kenza Sackho
- Department of Biochemical Sciences, University of Surrey, Guildford GU2 7XH, UK; (J.D.); (K.S.); (P.C.)
| | - Paola Campagnolo
- Department of Biochemical Sciences, University of Surrey, Guildford GU2 7XH, UK; (J.D.); (K.S.); (P.C.)
| | - Vlad Stolojan
- Advanced Technology Institute, School of Computer Science and Electronic Engineering, University of Surrey, Guildford GU2 7XH, UK; (C.J.H.); (A.P.)
| |
Collapse
|
10
|
Liu L, Tang H, Wang Y. Polymeric biomaterials: Advanced drug delivery systems in osteoarthritis treatment. Heliyon 2023; 9:e21544. [PMID: 38034809 PMCID: PMC10682535 DOI: 10.1016/j.heliyon.2023.e21544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Polymeric biomaterials have emerged as a highly promising candidate for drug delivery systems (DDS), exhibiting significant potential to enhance the therapeutic landscape of osteoarthritis (OA) therapy. Their remarkable capacity to manifest desirable physicochemical attributes, coupled with their excellent biocompatibility and biodegradability, has greatly expanded their utility in pharmacotherapeutic applications. Nevertheless, an urgent necessity exists for a comprehensive synthesis of the most recent advances in polymeric DDS, providing valuable guidance for their implementation in the context of OA therapy. This review is dedicated to summarizing and examining recent developments in the utilization of polymeric DDS for OA therapy. Initially, we present an overview of the intricate pathophysiology characterizing OA and underscore the prevailing limitations inherent to current treatment modalities. Subsequently, we introduce diverse categories of polymeric DDS, including hydrogels, nanofibers, and microspheres, elucidating their inherent advantages and limitations. Moreover, we discuss and summarize the delivery of bioactive agents through polymeric biomaterials for OA therapy, emphasizing key findings and emerging trends. Finally, we highlight prospective directions for advancing polymeric DDS, offering a promising approach to enhance their translational potential for OA therapy.
Collapse
Affiliation(s)
- Lin Liu
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| | - Haifeng Tang
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| | - Yanjun Wang
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| |
Collapse
|
11
|
Huang Y, Zhao H, Wang Y, Bi S, Zhou K, Li H, Zhou C, Wang Y, Wu W, Peng B, Tang J, Pan B, Wang B, Chen Z, Li Z, Zhang Z. The application and progress of tissue engineering and biomaterial scaffolds for total auricular reconstruction in microtia. Front Bioeng Biotechnol 2023; 11:1089031. [PMID: 37811379 PMCID: PMC10556751 DOI: 10.3389/fbioe.2023.1089031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/21/2023] [Indexed: 10/10/2023] Open
Abstract
Microtia is a congenital deformity of the ear with an incidence of about 0.8-4.2 per 10,000 births. Total auricular reconstruction is the preferred treatment of microtia at present, and one of the core technologies is the preparation of cartilage scaffolds. Autologous costal cartilage is recognized as the best material source for constructing scaffold platforms. However, costal cartilage harvest can lead to donor-site injuries such as pneumothorax, postoperative pain, chest wall scar and deformity. Therefore, with the need of alternative to autologous cartilage, in vitro and in vivo studies of biomaterial scaffolds and cartilage tissue engineering have gradually become novel research hot points in auricular reconstruction research. Tissue-engineered cartilage possesses obvious advantages including non-rejection, minimally invasive or non-invasive, the potential of large-scale production to ensure sufficient donors and controllable morphology. Exploration and advancements of tissue-engineered cartilaginous framework are also emerging in aspects including three-dimensional biomaterial scaffolds, acquisition of seed cells and chondrocytes, 3D printing techniques, inducing factors for chondrogenesis and so on, which has greatly promoted the research process of biomaterial substitute. This review discussed the development, current application and research progress of cartilage tissue engineering in auricular reconstruction, particularly the usage and creation of biomaterial scaffolds. The development and selection of various types of seed cells and inducing factors to stimulate chondrogenic differentiation in auricular cartilage were also highlighted. There are still confronted challenges before the clinical application becomes widely available for patients, and its long-term effect remains to be evaluated. We hope to provide guidance for future research directions of biomaterials as an alternative to autologous cartilage in ear reconstruction, and finally benefit the transformation and clinical application of cartilage tissue engineering and biomaterials in microtia treatment.
Collapse
Affiliation(s)
- Yeqian Huang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hanxing Zhao
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Yixi Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siwei Bi
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Zhou
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Hairui Li
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Yudong Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqing Wu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Peng
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Jun Tang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Bo Pan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baoyun Wang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhixing Chen
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Plastic Reconstructive and Aesthetic Surgery, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Zamora I, Alfonso Morales G, Castro JI, Ruiz Rojas LM, Valencia-Llano CH, Mina Hernandez JH, Valencia Zapata ME, Grande-Tovar CD. Chitosan (CS)/Hydroxyapatite (HA)/Tricalcium Phosphate (β-TCP)-Based Composites as a Potential Material for Pulp Tissue Regeneration. Polymers (Basel) 2023; 15:3213. [PMID: 37571109 PMCID: PMC10421191 DOI: 10.3390/polym15153213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
This research focused on developing new materials for endodontic treatments to restore tissues affected by infectious or inflammatory processes. Three materials were studied, namely tricalcium phosphate β-hydroxyapatite (β-TCP), commercial and natural hydroxyapatite (HA), and chitosan (CS), in different proportions. The chemical characterization using infrared spectroscopy (FTIR) and X-ray diffraction (XRD) analysis confirmed the composition of the composite. Scanning electron microscopy (SEM) demonstrated that the design and origin of the HA, whether natural or commercial, did not affect the morphology of the composites. In vitro studies using Artemia salina (A. salina) indicated that all three experimental materials were biocompatible after 24 h, with no significant differences in mortality rate observed among the groups. The subdermal implantation of the materials in block form exhibited biocompatibility and biodegradability after 30 and 60 days, with the larger particles undergoing fragmentation and connective tissue formation consisting of collagen type III fibers, blood vessels, and inflammatory cells. The implanted material continued to undergo resorption during this process. The results obtained in this research contribute to developing endodontic technologies for tissue recovery and regeneration.
Collapse
Affiliation(s)
- Ingrid Zamora
- Grupo Biomateriales Dentales, Escuela de Odontología, Universidad del Valle, Calle 4B # 36-00, Cali 76001, Colombia; (I.Z.); (G.A.M.); (C.H.V.-L.)
| | - Gilbert Alfonso Morales
- Grupo Biomateriales Dentales, Escuela de Odontología, Universidad del Valle, Calle 4B # 36-00, Cali 76001, Colombia; (I.Z.); (G.A.M.); (C.H.V.-L.)
| | - Jorge Iván Castro
- Laboratorio SIMERQO, Departamento de Química, Universidad del Valle, Calle 13 No. 100-00, Cali 76001, Colombia;
| | - Lina Marcela Ruiz Rojas
- Grupo de Materiales Compuestos, Escuela de Ingeniería de Materiales, Facultad de Ingeniería, Universidad del Valle, Calle 13 No. 100-00, Cali 760032, Colombia; (L.M.R.R.); (J.H.M.H.); (M.E.V.Z.)
| | - Carlos Humberto Valencia-Llano
- Grupo Biomateriales Dentales, Escuela de Odontología, Universidad del Valle, Calle 4B # 36-00, Cali 76001, Colombia; (I.Z.); (G.A.M.); (C.H.V.-L.)
| | - Jose Herminsul Mina Hernandez
- Grupo de Materiales Compuestos, Escuela de Ingeniería de Materiales, Facultad de Ingeniería, Universidad del Valle, Calle 13 No. 100-00, Cali 760032, Colombia; (L.M.R.R.); (J.H.M.H.); (M.E.V.Z.)
| | - Mayra Eliana Valencia Zapata
- Grupo de Materiales Compuestos, Escuela de Ingeniería de Materiales, Facultad de Ingeniería, Universidad del Valle, Calle 13 No. 100-00, Cali 760032, Colombia; (L.M.R.R.); (J.H.M.H.); (M.E.V.Z.)
| | - Carlos David Grande-Tovar
- Grupo de Investigación de Fotoquímica y Fotobiología, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| |
Collapse
|
13
|
Peng Y, Jiang H, Zuo HD. Factors affecting osteogenesis and chondrogenic differentiation of mesenchymal stem cells in osteoarthritis. World J Stem Cells 2023; 15:548-560. [PMID: 37424946 PMCID: PMC10324504 DOI: 10.4252/wjsc.v15.i6.548] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 06/26/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that often involves progressive cartilage degeneration and bone destruction of subchondral bone. At present, clinical treatment is mainly for pain relief, and there are no effective methods to delay the progression of the disease. When this disease progresses to the advanced stage, the only treatment option for most patients is total knee replacement surgery, which causes patients great pain and anxiety. As a type of stem cell, mesenchymal stem cells (MSCs) have multidirectional differentiation potential. The osteogenic differentiation and chondrogenic differentiation of MSCs can play vital roles in the treatment of OA, as they can relieve pain in patients and improve joint function. The differentiation direction of MSCs is accurately controlled by a variety of signaling pathways, so there are many factors that can affect the differentiation direction of MSCs by acting on these signaling pathways. When MSCs are applied to OA treatment, the microenvironment of the joints, injected drugs, scaffold materials, source of MSCs and other factors exert specific impacts on the differentiation direction of MSCs. This review aims to summarize the mechanisms by which these factors influence MSC differentiation to produce better curative effects when MSCs are applied clinically in the future.
Collapse
Affiliation(s)
- Yi Peng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Radiology, Chengdu Xinhua Hospital, Chengdu 610067, Sichuan Province, China
| |
Collapse
|
14
|
Bini F, D'Alessandro S, Pica A, Marinozzi F, Cidonio G. Harnessing Biofabrication Strategies to Re-Surface Osteochondral Defects: Repair, Enhance, and Regenerate. Biomimetics (Basel) 2023; 8:260. [PMID: 37366855 DOI: 10.3390/biomimetics8020260] [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: 05/24/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
Osteochondral tissue (OC) is a complex and multiphasic system comprising cartilage and subchondral bone. The discrete OC architecture is layered with specific zones characterized by different compositions, morphology, collagen orientation, and chondrocyte phenotypes. To date, the treatment of osteochondral defects (OCD) remains a major clinical challenge due to the low self-regenerative capacity of damaged skeletal tissue, as well as the critical lack of functional tissue substitutes. Current clinical approaches fail to fully regenerate damaged OC recapitulating the zonal structure while granting long-term stability. Thus, the development of new biomimetic treatment strategies for the functional repair of OCDs is urgently needed. Here, we review recent developments in the preclinical investigation of novel functional approaches for the resurfacing of skeletal defects. The most recent studies on preclinical augmentation of OCDs and highlights on novel studies for the in vivo replacement of diseased cartilage are presented.
Collapse
Affiliation(s)
- Fabiano Bini
- Department of Mechanical and Aerospace Engineering, Sapienza University, 00148 Rome, Italy
| | - Salvatore D'Alessandro
- Department of Mechanical and Aerospace Engineering, Sapienza University, 00148 Rome, Italy
- Center for Life Nano- & Neuro-Science (CLN2S), Fondazione Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Andrada Pica
- Department of Mechanical and Aerospace Engineering, Sapienza University, 00148 Rome, Italy
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Franco Marinozzi
- Department of Mechanical and Aerospace Engineering, Sapienza University, 00148 Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science (CLN2S), Fondazione Istituto Italiano di Tecnologia, 00161 Rome, Italy
| |
Collapse
|
15
|
Banihashemian A, Benisi SZ, Hosseinzadeh S, Shojaei S. Biomimetic biphasic scaffolds in osteochondral tissue engineering: Their composition, structure and consequences. Acta Histochem 2023; 125:152023. [PMID: 36940532 DOI: 10.1016/j.acthis.2023.152023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
Approaches to the design and construction of biomimetic scaffolds for osteochondral tissue, show increasing advances. Considering the limitations of this tissue in terms of repair and regeneration, there is a need to develop appropriately designed scaffolds. A combination of biodegradable polymers especially natural polymers and bioactive ceramics, shows promise in this field. Due to the complicated architecture of this tissue, biphasic and multiphasic scaffolds containing two or more different layers, could mimic the physiology and function of this tissue with a higher degree of similarity. The purpose of this review article is to discuss the approaches focused on the application of biphasic scaffolds for osteochondral tissue engineering, common methods of combining layers and the ultimate consequences of their use in patients were discussed.
Collapse
Affiliation(s)
- Abdolvahab Banihashemian
- Advanced Medical Sciences and Technologies Department, Faculty of Biomedical Engineering, Central Tehran Branch Islamic Azad University, Tehran, Iran.
| | - Soheila Zamanlui Benisi
- Stem Cell Research Center, Tissue Engineering and Regenerative Medicine Institute, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | - Simzar Hosseinzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shahrokh Shojaei
- Islamic Azad University Central Tehran Branch, Department of Biomedical Engineering, Tehran, Iran
| |
Collapse
|
16
|
Miri L, Irani S, Pezeshki-Modaress M, Daemi H, Atyabi SM. Guiding mesenchymal stem cells differentiation into chondrocytes using sulfated alginate/cold atmospheric plasma modified polycaprolactone nanofibrous scaffold. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04476-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
Wee AS, Lim CK, Tan SL, Ahmad TS, Kamarul T. TGF-β1 and -β3 for mesenchymal stem cells chondrogenic differentiation on PVA-NOCC-PEG scaffold. Tissue Eng Part C Methods 2022; 28:501-510. [PMID: 36082992 DOI: 10.1089/ten.tec.2022.0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) has been reported to promote chondrogenic differentiation and proliferation in the multipotent stromal cell (MSCs), and the transforming growth factor-beta 3 (TGF-β3) tends to be exclusively in promoting cell differentiation alone. The objective of this study was to determine the effect of TGF-β1 and -β3 on the MSCs chondrogenic differentiation on the poly (vinyl alcohol)-chitosan-poly (ethylene glycol) (PVA-NOCC-PEG) scaffold, compared to that of monolayer and pellet cultures. In this study, P2 rabbit bone marrow-derived MSCs were seeded either on the untreated 6-well plate (for monolayer culture) or onto the PVA-NOCC-PEG scaffold or cultured as a pellet culture. The cultures were maintained in a chemically defined serum-free medium supplemented with 10 ng/mL of either TGF-β1 or TGF-β3. Cell viability assay, biochemical assay and real-time polymerase chain reaction (qRT-PCR) were performed to determine the net effect of cell proliferation and chondrogenic differentiation of each of the growth factors. The results showed that the PVA-NOCC-PEG scaffold enhanced MSCs cell proliferation from day-12 to day-30 (p<0.05), however, no significant differences were observed in the cell proliferation between the cultures supplemented with or without TGF-β1 and TGF-β3 (p>0.05). In terms of chondrogenic differentiation, the PVA-NOCC-PEG scaffold augmented the GAGs secretion in MSCs and the mRNA expression levels of SOX9, type-II collagen, aggrecan and COMP were elevated (p<0.05). However, there was no significant difference between both the TGF-β1 and TGF-β3-treated groups (p>0.05). In conclusion, TGF-β1 and TGF-β3 enhanced the chondrogenic differentiation of MSCs seeded on the PVA-NOCC-PEG scaffold, however, there was no significant difference between the effect of TGF-β1 and TGF-β3.
Collapse
Affiliation(s)
- Ai-Sze Wee
- University of Malaya, Tissue Engineering Group (TEG), National Orthopaedics Centre of Excellent Research & Learning (NOCERAL), Department of Orthopaedic Surgery, Universiti Malaya, Lembah Pantai, Kuala Lumpur, Federal Territory, Malaysia.,SEGi University Kota Damansara, School of Allied Health Sciences, Petaling Jaya, Selangor, Malaysia;
| | - Chin-Keong Lim
- Skin Research and Innovation Centre, Research & Development Division, Wipro Manufacturing Services Sdn Bhd, Selangor, Subang Jaya, Malaysia;
| | - Sik Loo Tan
- University of Malaya, Department of Orthopaedic Surgery, Lembah Pantai, Kuala Lumpur, Federal Territory, Malaysia, 50603;
| | - Tunku Sara Ahmad
- University of Malaya, Department of Orthopaedic Surgery, Kuala Lumpur, Federal Territory, Malaysia;
| | - Tunku Kamarul
- University of Malaya, Tissue Engineering Group (TEG), National Orthopaedics Centre of Excellent Research & Learning (NOCERAL), Department of Orthopaedic Surgery, Universiti Malaya, Lembah Pantai, Kuala Lumpur, Federal Territory, Malaysia;
| |
Collapse
|
18
|
Miceli M, Maruotti GM, Sarno L, Carbone L, Guida M, Pelagalli A. Preliminary Characterization of the Epigenetic Modulation in the Human Mesenchymal Stem Cells during Chondrogenic Process. Int J Mol Sci 2022; 23:ijms23179870. [PMID: 36077266 PMCID: PMC9456537 DOI: 10.3390/ijms23179870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Regenerative medicine represents a growing hot topic in biomedical sciences, aiming at setting out novel therapeutic strategies to repair or regenerate damaged tissues and organs. For this perspective, human mesenchymal stem cells (hMSCs) play a key role in tissue regeneration, having the potential to differentiate into many cell types, including chondrocytes. Accordingly, in the last few years, researchers have focused on several in vitro strategies to optimize hMSC differentiation protocols, including those relying on epigenetic manipulations that, in turn, lead to the modulation of gene expression patterns. Therefore, in the present study, we investigated the role of the class II histone deacetylase (HDAC) inhibitor, MC1568, in the hMSCs-derived chondrogenesis. The hMSCs we used for this work were the hMSCs obtained from the amniotic fluid, given their greater differentiation capacity. Our preliminary data documented that MC1568 drove both the improvement and acceleration of hMSCs chondrogenic differentiation in vitro, since the differentiation process in MC1568-treated cells took place in about seven days, much less than that normally observed, namely 21 days. Collectively, these preliminary data might shed light on the validity of such a new differentiative protocol, in order to better assess the potential role of the epigenetic modulation in the process of the hypertrophic cartilage formation, which represents the starting point for endochondral ossification.
Collapse
Affiliation(s)
- Marco Miceli
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy
- Correspondence: (M.M.); (A.P.)
| | - Giuseppe Maria Maruotti
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy
| | - Laura Sarno
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy
| | - Luigi Carbone
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy
| | - Maurizio Guida
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), 80131 Naples, Italy
- Correspondence: (M.M.); (A.P.)
| |
Collapse
|
19
|
Nano-Structured Ridged Micro-Filaments (≥100 µm Diameter) Produced Using a Single Step Strategy for Improved Bone Cell Adhesion and Proliferation in Textile Scaffolds. Molecules 2022; 27:molecules27123790. [PMID: 35744916 PMCID: PMC9228432 DOI: 10.3390/molecules27123790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 12/01/2022] Open
Abstract
Textile scaffolds that are either 2D or 3D with tunable shapes and pore sizes can be made through textile processing (weaving, knitting, braiding, nonwovens) using microfilaments. However, these filaments lack nano-topographical features to improve bone cell adhesion and proliferation. Moreover, the diameter of such filaments should be higher than that used for classical textiles (10−30 µm) to enable adhesion and the efficient spreading of the osteoblast cell (>30 µm diameter). We report, for the first time, the fabrication of biodegradable nanostructured cylindrical PLLA (poly-L-Lactic acid) microfilaments of diameters 100 µm and 230 µm, using a single step melt-spinning process for straightforward integration of nano-scale ridge-like structures oriented in the fiber length direction. Appropriate drawing speed and temperature used during the filament spinning allowed for the creation of instabilities giving rise to nanofibrillar ridges, as observed by AFM (Atomic Force Microscopy). These micro-filaments were hydrophobic, and had reduced crystallinity and mechanical strength, but could still be processed into 2D/3D textile scaffolds of various shapes. Biological tests carried out on the woven scaffolds made from these nano-structured micro filaments showed excellent human bone cell MG 63 adhesion and proliferation, better than on smooth 30 µm- diameter fibers. Elongated filopodia of the osteoblast, intimately anchored to the nano-structured filaments, was observed. The filaments also induced in vitro osteogenic expression, as shown by the expression of osteocalcin and bone sialoprotein after 21 days of culture. This work deals with the fabrication of a new generation of nano-structured micro-filament for use as scaffolds of different shapes suited for bone cell engineering.
Collapse
|
20
|
Owida HA, Al-Nabulsi JI, Alnaimat F, Al-Ayyad M, Turab NM, Al Sharah A, Shakur M. Recent Applications of Electrospun Nanofibrous Scaffold in Tissue Engineering. Appl Bionics Biomech 2022; 2022:1953861. [PMID: 35186119 PMCID: PMC8849965 DOI: 10.1155/2022/1953861] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/12/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering is a relatively new area of research that combines medical, biological, and engineering fundamentals to create tissue-engineered constructs that regenerate, preserve, or slightly increase the functions of tissues. To create mature tissue, the extracellular matrix should be imitated by engineered structures, allow for oxygen and nutrient transmission, and release toxins during tissue repair. Numerous recent studies have been devoted to developing three-dimensional nanostructures for tissue engineering. One of the most effective of these methods is electrospinning. Numerous nanofibrous scaffolds have been constructed over the last few decades for tissue repair and restoration. The current review gives an overview of attempts to construct nanofibrous meshes as tissue-engineered scaffolds for various tissues such as bone, cartilage, cardiovascular, and skin tissues. Also, the current article addresses the recent improvements and difficulties in tissue regeneration using electrospinning.
Collapse
Affiliation(s)
- Hamza Abu Owida
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Jamal I. Al-Nabulsi
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Feras Alnaimat
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Muhammad Al-Ayyad
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Nidal M. Turab
- Department of Networks and Information Security, Faculty of Information Technology, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Ashraf Al Sharah
- Computer Engineering, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Murad Shakur
- Medical Engineering Department, Faculty of Engineering, Al-Ahliyya Amman University, Amman 19328, Jordan
| |
Collapse
|
21
|
Yamada K, Iwasaki N, Sudo H. Biomaterials and Cell-Based Regenerative Therapies for Intervertebral Disc Degeneration with a Focus on Biological and Biomechanical Functional Repair: Targeting Treatments for Disc Herniation. Cells 2022; 11:602. [PMID: 35203253 PMCID: PMC8870062 DOI: 10.3390/cells11040602] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/22/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a common cause of low back pain and most spinal disorders. As IVD degeneration is a major obstacle to the healthy life of so many individuals, it is a major issue that needs to be overcome. Currently, there is no clinical treatment for the regeneration of degenerated IVDs. However, recent advances in regenerative medicine and tissue engineering suggest the potential of cell-based and/or biomaterial-based IVD regeneration therapies. These treatments may be indicated for patients with IVDs in the intermediate degenerative stage, a point where the number of viable cells decreases, and the structural integrity of the disc begins to collapse. However, there are many biological, biomechanical, and clinical challenges that must be overcome before the clinical application of these IVD regeneration therapies can be realized. This review summarizes the basic research and clinical trials literature on cell-based and biomaterial-based IVD regenerative therapies and outlines the important role of these strategies in regenerative treatment for IVD degenerative diseases, especially disc herniation.
Collapse
Affiliation(s)
- Katsuhisa Yamada
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (K.Y.); (N.I.)
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (K.Y.); (N.I.)
| | - Hideki Sudo
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
22
|
Sun K, Tao C, Wang DA. Scaffold-free approaches for the fabrication of engineered articular cartilage tissue. Biomed Mater 2022; 17. [PMID: 35114657 DOI: 10.1088/1748-605x/ac51b9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/03/2022] [Indexed: 11/12/2022]
Abstract
Tissue engineered cartilaginous constructs have meet great advances in the past decades as a treatment for osteoarthritis, a degenerative disease affecting people all over the world as the population ages. Scaffold-free tissue engineered constructs are designed and developed in recent years with only cells and cell-derived matrix involved. Scaffold-free tissue constructs do not require cell adherence on exogenous materials and are superior to scaffold-based constructs in (1) relying on only cells to produce matrix, (2) not interfering cell-cell signaling, cell migration or small molecules diffusion after implantation and (3) introducing no exogenous impurities. In this review, three main scaffold-free methodologies for cartilage tissue engineering, the cell sheet technology, the phase transfer cell culture-living hyaline cartilage graft (PTCC-LhCG) system and the cell aggregate-based (bottom-up) methods, were reviewed, covering mold fabrication, decellularization and 3D bioprinting. The recent advances, medical applications, superiority and drawbacks were elaborated in detail.
Collapse
Affiliation(s)
- Kang Sun
- City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Kowloon, 000000, HONG KONG
| | - Chao Tao
- City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Kowloon, 000000, HONG KONG
| | - Dong-An Wang
- City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Kowloon, 000000, HONG KONG
| |
Collapse
|
23
|
Wang S, Xu J, Li W, Sun S, Gao S, Hou Y. Magnetic Nanostructures: Rational Design and Fabrication Strategies toward Diverse Applications. Chem Rev 2022; 122:5411-5475. [PMID: 35014799 DOI: 10.1021/acs.chemrev.1c00370] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, the continuous development of magnetic nanostructures (MNSs) has tremendously promoted both fundamental scientific research and technological applications. Different from the bulk magnet, the systematic engineering on MNSs has brought a great breakthrough in some emerging fields such as the construction of MNSs, the magnetism exploration of multidimensional MNSs, and their potential translational applications. In this review, we give a detailed description of the synthetic strategies of MNSs based on the fundamental features and application potential of MNSs and discuss the recent progress of MNSs in the fields of nanomedicines, advanced nanobiotechnology, catalysis, and electromagnetic wave adsorption (EMWA), aiming to provide guidance for fabrication strategies of MNSs toward diverse applications.
Collapse
Affiliation(s)
- Shuren Wang
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Junjie Xu
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Wei Li
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Shengnan Sun
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Song Gao
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Institute of Spin-X Science and Technology, South China University of Technology, Guangzhou 511442, China
| | - Yanglong Hou
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| |
Collapse
|
24
|
Sustainable Applications of Nanofibers in Agriculture and Water Treatment: A Review. SUSTAINABILITY 2022. [DOI: 10.3390/su14010464] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Natural fibers are an important source for producing polymers, which are highly applicable in their nanoform and could be used in very broad fields such as filtration for water/wastewater treatment, biomedicine, food packaging, harvesting, and storage of energy due to their high specific surface area. These natural nanofibers could be mainly produced through plants, animals, and minerals, as well as produced from agricultural wastes. For strengthening these natural fibers, they may reinforce with some substances such as nanomaterials. Natural or biofiber-reinforced bio-composites and nano–bio-composites are considered better than conventional composites. The sustainable application of nanofibers in agricultural sectors is a promising approach and may involve plant protection and its growth through encapsulating many bio-active molecules or agrochemicals (i.e., pesticides, phytohormones, and fertilizers) for smart delivery at the targeted sites. The food industry and processing also are very important applicable fields of nanofibers, particularly food packaging, which may include using nanofibers for active–intelligent food packaging, and food freshness indicators. The removal of pollutants from soil, water, and air is an urgent field for nanofibers due to their high efficiency. Many new approaches or applicable agro-fields for nanofibers are expected in the future, such as using nanofibers as the indicators for CO and NH3. The role of nanofibers in the global fighting against COVID-19 may represent a crucial solution, particularly in producing face masks.
Collapse
|
25
|
Functional Graphene Nanomaterials-Based Hybrid Scaffolds for Osteogenesis and Chondrogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1351:65-87. [DOI: 10.1007/978-981-16-4923-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Karacan I, Milthorpe B, Ben-Nissan B, Santos J. Stem Cells and Proteomics in Biomaterials and Biomedical Applications. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2022:125-157. [DOI: 10.1007/978-981-16-7435-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
27
|
Li C, Chen J, Lv Y, Liu Y, Guo Q, Wang J, Wang C, Hu P, Liu Y. Recent Progress in Electrospun Nanofiber-Based Degenerated Intervertebral Disc Repair. ACS Biomater Sci Eng 2021; 8:16-31. [PMID: 34913688 DOI: 10.1021/acsbiomaterials.1c00970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Annulus fibrosus fissure and fibrosis of nucleus pulposus are severe morphological characteristics of intervertebral disc degeneration. Currently, surgery or drugs are used to relieve pain in such cases. Tissue engineering is a new multidisciplinary strategy with great potential for use in joint replacement and organ regeneration. Based on the natural anatomy of intervertebral discs, intervertebral disc scaffolds are fabricated by exploiting the special arrangement of extracellular matrix fibers. Electrospun nanofibers possess clear advantages in repairing degenerated intervertebral discs. This article reviews and summarizes recently developed methods for improving and fabricating electrospun nanofiber annulus fibrosus scaffolds in terms of nanofiber alignment, material selection, loading additives, and the progress made in combining other advanced technologies with electrospun nanofibers. In addition, the improvement in mechanical properties and biocompatibility of nucleus pulposus scaffolds by electrospun nanofiber-reinforced hydrogels is discussed. Finally, complete intervertebral disc scaffolds can be fabricated using the disc-like angle-ply structure and other emerging fabrication methods. Taken together, electrospun nanofiber intervertebral disc scaffolds are promising for clinical applications.
Collapse
Affiliation(s)
- Chenxi Li
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jia Chen
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yarong Lv
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yueqi Liu
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Quanyi Guo
- Institute of Orthopedics, the Fourth Medical Center, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiandong Wang
- Division of Breast Surgery, Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ce Wang
- Alan G. MacDiarmid Institute, Jilin University, Changchun, Jilin 130012, China
| | - Ping Hu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yong Liu
- Beijing Key Laboratory of Advanced Functional Polymer Composites, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
28
|
Mandal S, Dube T, Mohapatra AK, Choudhury S, Khanam F, Yadav P, Chauhan VS, Mishra J, Panda JJ. Engineered Biocompatible and Stable Dipeptide Hydrogel with Tunable Mechanical and Cell Growth Properties to Embolden Neuroglial Cell Growth. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10290-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
29
|
Combinations of Hydrogels and Mesenchymal Stromal Cells (MSCs) for Cartilage Tissue Engineering-A Review of the Literature. Gels 2021; 7:gels7040217. [PMID: 34842678 PMCID: PMC8628761 DOI: 10.3390/gels7040217] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/11/2021] [Accepted: 11/13/2021] [Indexed: 01/17/2023] Open
Abstract
Cartilage offers limited regenerative capacity. Cell-based approaches have emerged as a promising alternative in the treatment of cartilage defects and osteoarthritis. Due to their easy accessibility, abundancy, and chondrogenic potential mesenchymal stromal cells (MSCs) offer an attractive cell source. MSCs are often combined with natural or synthetic hydrogels providing tunable biocompatibility, biodegradability, and enhanced cell functionality. In this review, we focused on the different advantages and disadvantages of various natural, synthetic, and modified hydrogels. We examined the different combinations of MSC-subpopulations and hydrogels used for cartilage engineering in preclinical and clinical studies and reviewed the effects of added growth factors or gene transfer on chondrogenesis in MSC-laden hydrogels. The aim of this review is to add to the understanding of the disadvantages and advantages of various combinations of MSC-subpopulations, growth factors, gene transfers, and hydrogels in cartilage engineering.
Collapse
|
30
|
Zhuang Y, Cui W. Biomaterial-based delivery of nucleic acids for tissue regeneration. Adv Drug Deliv Rev 2021; 176:113885. [PMID: 34324886 DOI: 10.1016/j.addr.2021.113885] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022]
Abstract
Gene therapy is a promising novel method of tissue regeneration by stimulating or inhibiting key signaling pathways. However, their therapeutic applications in vivo are largely limited by several physiological obstacles, such as degradation of nucleases, impermeability of cell membranes, and transport to the desired intracellular compartments. Biomaterial-based gene delivery systems can overcome the problems of stability and local drug delivery, and can temporarily control the overexpression of therapeutic genes, leading to the local production of physiologically relevant levels of regulatory factors. But the gene delivery of biomaterials for tissue regeneration relies on multi-factor design. This review aims to outline the impact of gene delivery methods, therapeutic genes and biomaterials selection on this strategy, emphatically introduce the latest developments in the design of gene delivery vehicles based on biomaterials, summarize the mechanism of nucleic acid for tissue regeneration, and explore the strategies of nucleic acid delivery vehicles for various tissue regeneration.
Collapse
Affiliation(s)
- Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention, Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention, Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
31
|
Idumah CI, Ezika AC. Recent advancements in hybridized polymer nano-biocomposites for tissue engineering. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1960344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Christopher Igwe Idumah
- Department of Polymer and Textile Engineering, Faculty of Engineering, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Anthony Chidi Ezika
- Institute of NanoEngineering Research (INER) and Department of Chemical, Metallurgical and Materials Engineering, Faculty of Engineering and The Built Environment, Tshwane University of Technology, Pretoria, South Africa
| |
Collapse
|
32
|
Kamoun EA, Loutfy SA, Hussein Y, Kenawy ERS. Recent advances in PVA-polysaccharide based hydrogels and electrospun nanofibers in biomedical applications: A review. Int J Biol Macromol 2021; 187:755-768. [PMID: 34358597 DOI: 10.1016/j.ijbiomac.2021.08.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/22/2021] [Accepted: 08/01/2021] [Indexed: 02/08/2023]
Abstract
Among several types of carbohydrate polymers blend PVA hydrogel membranes used for biomedical applications in particular wound dressings; electrospun nanofibrous membranes have gained increased interest because of their extraordinary features e.g. huge surface area to volume ratio, high porosity, adequate permeability, excellent wound-exudates absorption capacity, architecture similarity with skin ECM and sustained release-profile over long time. In this study, modern perspectives of synthesized/developed electrospun nanofibrous hydrogel membranes based popular carbohydrate polymers blend PVA which recently have been employed for versatile biomedical applications particularly wound dressings, were discussed intensively and compared in detail with traditional fabricated membranes based films, as well. Clinically relevant and advantages of electrospun nanofibrous membranes were discussed in terms of their biocompatibility and easily fabrication and functionalization in different biomedical applications.
Collapse
Affiliation(s)
- Elbadawy A Kamoun
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE), El-Sherouk City, Cairo 11837, Egypt; Polymeric Materials Research Dep., Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab City 21934, Alexandria, Egypt.
| | - Samah A Loutfy
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE), El-Sherouk City, Cairo 11837, Egypt; Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Egypt
| | - Yasmein Hussein
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE), El-Sherouk City, Cairo 11837, Egypt
| | - El-Refaie S Kenawy
- Polymer Research Group, Department of Chemistry, Faculty of Science, University of Tanta, Tanta 31527, Egypt
| |
Collapse
|
33
|
Natarajan ABMT, Sivadas VPD, Nair PDPD. 3D-printed biphasic scaffolds for the simultaneous regeneration of osteochondral tissues. Biomed Mater 2021; 16. [PMID: 34265754 DOI: 10.1088/1748-605x/ac14cb] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/15/2021] [Indexed: 12/19/2022]
Abstract
Osteochondral tissue engineering (OCTE) involves the simulation of highly complex tissues with disparate biomechanical properties. OCTE is regarded as the best option for treating osteochondral defects, most of the drawbacks of current treatment methodologies can be addressed by this method. In recent years, the conventional scaffolds used in cartilage and bone regeneration are gradually being replaced by 3D printed scaffolds (3DP). In the present study, we devised the strategy of 3D printing for fabricating biphasic and integrated scaffolds that are loaded with bioactive factors for enhancing the osteochondral tissue regeneration. Polycaprolactone (PCL) and poly(lactic-co-glycolic acid) (PLGA), is used along with bioactive factors (chondroitin sulphate and beta-tricalcium phosphate (βTCP)) for the upper cartilage and lower bone layer respectively. The 3D printed bi-layered scaffolds with varying infill density, to mimic the native tissue, are not previously explored for OCTE. Hence, we tested the simultaneous osteochondrogenic differentiation inducing potential of the aforesaid 3D printed biphasic scaffoldsin vitro, using rabbit adipose derived mesenchymal stem cells (ADMSCs). Further, the biphasic scaffolds were highly cytocompatible, with excellent cell adhesion properties and cellular morphology. Most importantly, these biphasic scaffolds directed the simultaneous differentiation of a single stem cell population in to two cell lineages (simultaneous differentiation of rabbit ADMSCs into chondrocytes and osteoblasts). Further, these scaffolds enhanced the production of ECM and induced robust expression of marker genes that is specific for respective cartilage and bone layers. The 3D printed OCTE scaffold of our study hence can simulate the native osteochondral unit and could be potential futuristic biomimetic scaffold for osteochondral defects. Furtherin vivostudies are warranted.
Collapse
Affiliation(s)
- Amrita Bds MTech Natarajan
- Division of Tissue Engineering and Regeneration Technologies, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute of Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - Vp Ph D Sivadas
- Division of Tissue Engineering and Regeneration Technologies, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute of Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| | - Prabha D Ph D Nair
- Division of Tissue Engineering and Regeneration Technologies, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute of Medical Sciences and Technology, Thiruvananthapuram, Kerala 695012, India
| |
Collapse
|
34
|
Kumar R, Gulia K. The convergence of nanotechnology‐stem cell, nanotopography‐mechanobiology, and biotic‐abiotic interfaces: Nanoscale tools for tackling the top killer, arteriosclerosis, strokes, and heart attacks. NANO SELECT 2021. [DOI: 10.1002/nano.202000192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Rajiv Kumar
- NIET National Institute of Medical Science Rajasthan India
| | - Kiran Gulia
- Materials and Manufacturing School of Engineering University of Wolverhampton Wolverhampton England, UK
| |
Collapse
|
35
|
Li J, Liu Y, Zhang Y, Yao B, Enhejirigala, Li Z, Song W, Wang Y, Duan X, Yuan X, Fu X, Huang S. Biophysical and Biochemical Cues of Biomaterials Guide Mesenchymal Stem Cell Behaviors. Front Cell Dev Biol 2021; 9:640388. [PMID: 33842464 PMCID: PMC8027358 DOI: 10.3389/fcell.2021.640388] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely used in the fields of tissue engineering and regenerative medicine due to their self-renewal capabilities and multipotential differentiation assurance. However, capitalizing on specific factors to precisely guide MSC behaviors is the cornerstone of biomedical applications. Fortunately, several key biophysical and biochemical cues of biomaterials that can synergistically regulate cell behavior have paved the way for the development of cell-instructive biomaterials that serve as delivery vehicles for promoting MSC application prospects. Therefore, the identification of these cues in guiding MSC behavior, including cell migration, proliferation, and differentiation, may be of particular importance for better clinical performance. This review focuses on providing a comprehensive and systematic understanding of biophysical and biochemical cues, as well as the strategic engineering of these signals in current scaffold designs, and we believe that integrating biophysical and biochemical cues in next-generation biomaterials would potentially help functionally regulate MSCs for diverse applications in regenerative medicine and cell therapy in the future.
Collapse
Affiliation(s)
- Jianjun Li
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- Department of General Surgery, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yufan Liu
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
| | - Yijie Zhang
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
| | - Bin Yao
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Enhejirigala
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- College of Graduate, Tianjin Medical University, Tianjin, China
- Institute of Basic Medical Research, Inner Mongolia Medical University, Hohhot, China
| | - Zhao Li
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
| | - Wei Song
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
| | - Yuzhen Wang
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- Department of Burn and Plastic Surgery, Air Force Hospital of Chinese PLA Central Theater Command, Datong, China
| | - Xianlan Duan
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xingyu Yuan
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Chinese PLA General Hospital, PLA Medical College, Beijing, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration, Medical Innovation Research Department and the Fourth Medical Center, Chinese PLA General Hospital, PLA Medical College, Beijing, China
| |
Collapse
|
36
|
Theodoridis K, Manthou ME, Aggelidou E, Kritis A. In Vivo Cartilage Regeneration with Cell-Seeded Natural Biomaterial Scaffold Implants: 15-Year Study. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:206-245. [PMID: 33470169 DOI: 10.1089/ten.teb.2020.0295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Articular cartilage can be easily damaged from human's daily activities, leading to inflammation and to osteoarthritis, a situation that can diminish the patients' quality of life. For larger cartilage defects, scaffolds are employed to provide cells the appropriate three-dimensional environment to proliferate and differentiate into healthy cartilage tissue. Natural biomaterials used as scaffolds, attract researchers' interest because of their relative nontoxic nature, their abundance as natural products, their easy combination with other materials, and the relative easiness to establish Marketing Authorization. The last 15 years were chosen to review, document, and elucidate the developments on cell-seeded natural biomaterials for articular cartilage treatment in vivo. The parameters of the experimental designs and their results were all documented and presented. Considerations about the newly formed cartilage and the treatment of cartilage defects were discussed, along with difficulties arising when applying natural materials, research limitations, and tissue engineering approaches for hyaline cartilage regeneration.
Collapse
Affiliation(s)
- Konstantinos Theodoridis
- Department of Physiology and Pharmacology, Faculty of Health Sciences and cGMP Regenerative Medicine Facility, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Maria Eleni Manthou
- Laboratory of Histology, Embryology, and Anthropology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, Faculty of Health Sciences and cGMP Regenerative Medicine Facility, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, Faculty of Health Sciences and cGMP Regenerative Medicine Facility, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| |
Collapse
|
37
|
Lawson TB, Mäkelä JTA, Klein T, Snyder BD, Grinstaff MW. Nanotechnology and Osteoarthritis. Part 2: Opportunities for advanced devices and therapeutics. J Orthop Res 2021; 39:473-484. [PMID: 32860444 DOI: 10.1002/jor.24842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/27/2020] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) is a multifactorial disease of the entire joint which afflicts 140 million individuals worldwide regardless of economic or social status. Current clinical treatments for OA primarily center on reducing pain and increasing mobility, and there are limited therapeutic interventions to restore degraded cartilage or slow disease pathogenesis. This second installment of a two-part review on nanotechnology and OA focuses on novel treatment strategies. Specifically, Part 2 first discusses current surgical and nonsurgical treatments for OA and then summarizes recent advancements in nanotechnology-based treatments, while Part 1 (10.1002/jor.24817) described advances in imaging and diagnostics. We review nano delivery systems for small molecule drugs, nucleic acids, and proteins followed by nano-based scaffolds for neocartilage formation and osteochondral regeneration, and lastly nanoparticle lubricants. We conclude by identifying opportunities for nanomedicine advances, and prospects for OA treatments.
Collapse
Affiliation(s)
- Taylor B Lawson
- Departments of Biomedical Engineering, Mechanical Engineering, Chemistry, and Medicine Boston University, Boston, Massachusetts, USA
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Janne T A Mäkelä
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Travis Klein
- Center for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Brian D Snyder
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Mark W Grinstaff
- Departments of Biomedical Engineering, Mechanical Engineering, Chemistry, and Medicine Boston University, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Field J, Haycock JW, Boissonade FM, Claeyssens F. A Tuneable, Photocurable, Poly(Caprolactone)-Based Resin for Tissue Engineering-Synthesis, Characterisation and Use in Stereolithography. Molecules 2021; 26:1199. [PMID: 33668087 PMCID: PMC7956195 DOI: 10.3390/molecules26051199] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 11/16/2022] Open
Abstract
Stereolithography is a useful additive manufacturing technique for the production of scaffolds for tissue engineering. Here we present a tuneable, easy-to-manufacture, photocurable resin for use in stereolithography, based on the widely used biomaterial, poly(caprolactone) (PCL). PCL triol was methacrylated to varying degrees and mixed with photoinitiator to produce a photocurable prepolymer resin, which cured under UV light to produce a cytocompatible material. This study demonstrates that poly(caprolactone) methacrylate (PCLMA) can be produced with a range of mechanical properties and degradation rates. By increasing the degree of methacrylation (DM) of the prepolymer, the Young's modulus of the crosslinked PCLMA could be varied from 0.12-3.51 MPa. The accelerated degradation rate was also reduced from complete degradation in 17 days to non-significant degradation in 21 days. The additive manufacturing capabilities of the resin were demonstrated by the production of a variety of different 3D structures using micro-stereolithography. Here, β-carotene was used as a novel, cytocompatible photoabsorber and enabled the production of complex geometries by giving control over cure depth. The PCLMA presented here offers an attractive, tuneable biomaterial for the production of tissue engineering scaffolds for a wide range of applications.
Collapse
Affiliation(s)
- Jonathan Field
- The School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK; (J.F.); (F.M.B.)
| | - John W. Haycock
- The Department of Materials Science and Engineering, The University of Sheffield, Sheffield S3 7HQ, UK;
- The Neuroscience Institute, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Fiona M. Boissonade
- The School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK; (J.F.); (F.M.B.)
- The Neuroscience Institute, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Frederik Claeyssens
- The Department of Materials Science and Engineering, The University of Sheffield, Sheffield S3 7HQ, UK;
- The Neuroscience Institute, The University of Sheffield, Sheffield S10 2HQ, UK
| |
Collapse
|
39
|
Seaberg J, Montazerian H, Hossen MN, Bhattacharya R, Khademhosseini A, Mukherjee P. Hybrid Nanosystems for Biomedical Applications. ACS NANO 2021; 15:2099-2142. [PMID: 33497197 PMCID: PMC9521743 DOI: 10.1021/acsnano.0c09382] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Inorganic/organic hybrid nanosystems have been increasingly developed for their versatility and efficacy at overcoming obstacles not readily surmounted by nonhybridized counterparts. Currently, hybrid nanosystems are implemented for gene therapy, drug delivery, and phototherapy in addition to tissue regeneration, vaccines, antibacterials, biomolecule detection, imaging probes, and theranostics. Though diverse, these nanosystems can be classified according to foundational inorganic/organic components, accessory moieties, and architecture of hybridization. Within this Review, we begin by providing a historical context for the development of biomedical hybrid nanosystems before describing the properties, synthesis, and characterization of their component building blocks. Afterward, we introduce the architectures of hybridization and highlight recent biomedical nanosystem developments by area of application, emphasizing hybrids of distinctive utility and innovation. Finally, we draw attention to ongoing clinical trials before recapping our discussion of hybrid nanosystems and providing a perspective on the future of the field.
Collapse
Affiliation(s)
- Joshua Seaberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
| | - Hossein Montazerian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Md Nazir Hossen
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
40
|
Wong KU, Zhang A, Akhavan B, Bilek MM, Yeo GC. Biomimetic Culture Strategies for the Clinical Expansion of Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2021. [PMID: 33599471 DOI: 10.1021/acsbiomaterials.0c01538] [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
Mesenchymal stromal/stem cells (MSCs) typically require significant ex vivo expansion to achieve the high cell numbers required for research and clinical applications. However, conventional MSC culture on planar (2D) plastic surfaces has been shown to induce MSC senescence and decrease cell functionality over long-term proliferation, and usually, it has a high labor requirement, a high usage of reagents, and therefore, a high cost. In this Review, we describe current MSC-based therapeutic strategies and outline the important factors that need to be considered when developing next-generation cell expansion platforms. To retain the functional value of expanded MSCs, ex vivo culture systems should ideally recapitulate the components of the native stem cell microenvironment, which include soluble cues, resident cells, and the extracellular matrix substrate. We review the interplay between these stem cell niche components and their biological roles in governing MSC phenotype and functionality. We discuss current biomimetic strategies of incorporating biochemical and biophysical cues in MSC culture platforms to grow clinically relevant cell numbers while preserving cell potency and stemness. This Review summarizes the current state of MSC expansion technologies and the challenges that still need to be overcome for MSC clinical applications to be feasible and sustainable.
Collapse
Affiliation(s)
- Kuan Un Wong
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anyu Zhang
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Behnam Akhavan
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Marcela M Bilek
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Giselle C Yeo
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
41
|
Chocarro‐Wrona C, de Vicente J, Antich C, Jiménez G, Martínez‐Moreno D, Carrillo E, Montañez E, Gálvez‐Martín P, Perán M, López‐Ruiz E, Marchal JA. Validation of the 1,4-butanediol thermoplastic polyurethane as a novel material for 3D bioprinting applications. Bioeng Transl Med 2021; 6:e10192. [PMID: 33532591 PMCID: PMC7823129 DOI: 10.1002/btm2.10192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022] Open
Abstract
Tissue engineering (TE) seeks to fabricate implants that mimic the mechanical strength, structure, and composition of native tissues. Cartilage TE requires the development of functional personalized implants with cartilage-like mechanical properties capable of sustaining high load-bearing environments to integrate into the surrounding tissue of the cartilage defect. In this study, we evaluated the novel 1,4-butanediol thermoplastic polyurethane elastomer (b-TPUe) derivative filament as a 3D bioprinting material with application in cartilage TE. The mechanical behavior of b-TPUe in terms of friction and elasticity were examined and compared with human articular cartilage, PCL, and PLA. Moreover, infrapatellar fat pad-derived human mesenchymal stem cells (MSCs) were bioprinted together with scaffolds. in vitro cytotoxicity, proliferative potential, cell viability, and chondrogenic differentiation were analyzed by Alamar blue assay, SEM, confocal microscopy, and RT-qPCR. Moreover, in vivo biocompatibility and host integration were analyzed. b-TPUe demonstrated a much closer compression and shear behavior to native cartilage than PCL and PLA, as well as closer tribological properties to cartilage. Moreover, b-TPUe bioprinted scaffolds were able to maintain proper proliferative potential, cell viability, and supported MSCs chondrogenesis. Finally, in vivo studies revealed no toxic effects 21 days after scaffolds implantation, extracellular matrix deposition and integration within the surrounding tissue. This is the first study that validates the biocompatibility of b-TPUe for 3D bioprinting. Our findings indicate that this biomaterial can be exploited for the automated biofabrication of artificial tissues with tailorable mechanical properties including the great potential for cartilage TE applications.
Collapse
Affiliation(s)
- Carlos Chocarro‐Wrona
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Juan de Vicente
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
- Department of Applied PhysicsFaculty of Sciences, University of GranadaGranadaSpain
| | - Cristina Antich
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Gema Jiménez
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Daniel Martínez‐Moreno
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Esmeralda Carrillo
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| | - Elvira Montañez
- Biomedical Research Institute of Málaga (IBIMA)Málaga
- Department of Orthopedic Surgery and TraumatologyVirgen de la Victoria University HospitalMálagaSpain
| | - Patricia Gálvez‐Martín
- Department of Pharmacy and Pharmaceutical TechnologySchool of Pharmacy, University of GranadaGranadaSpain
- Advanced Therapies AreaBioibérica S.A.UBarcelonaSpain
| | - Macarena Perán
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
- Department of Health SciencesUniversity of JaénJaénSpain
| | - Elena López‐Ruiz
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
- Department of Health SciencesUniversity of JaénJaénSpain
| | - Juan Antonio Marchal
- Biosanitary Research Institute of Granada (ibs.GRANADA)University Hospitals of Granada‐University of GranadaGranadaSpain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of GranadaGranadaSpain
- Department of Human Anatomy and EmbryologyFaculty of Medicine, University of GranadaGranadaSpain
- Excellence Research Unit “Modeling Nature” (MNat)University of GranadaGranadaSpain
| |
Collapse
|
42
|
Vasanthan J, Gurusamy N, Rajasingh S, Sigamani V, Kirankumar S, Thomas EL, Rajasingh J. Role of Human Mesenchymal Stem Cells in Regenerative Therapy. Cells 2020; 10:E54. [PMID: 33396426 PMCID: PMC7823630 DOI: 10.3390/cells10010054] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells which can proliferate and replace dead cells in the body. MSCs also secrete immunomodulatory molecules, creating a regenerative microenvironment that has an excellent potential for tissue regeneration. MSCs can be easily isolated and grown in vitro for various applications. For the past two decades, MSCs have been used in research, and many assays and tests have been developed proving that MSCs are an excellent cell source for therapy. This review focusses on quality control parameters required for applications of MSCs including colony formation, surface markers, differentiation potentials, and telomere length. Further, the specific mechanisms of action of MSCs under various conditions such as trans-differentiation, cell fusion, mitochondrial transfer, and secretion of extracellular vesicles are discussed. This review aims to underline the applications and benefits of MSCs in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Edwin L. Thomas
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
43
|
Yu LM, Liu T, Ma YL, Zhang F, Huang YC, Fan ZH. Fabrication of Silk-Hyaluronan Composite as a Potential Scaffold for Tissue Repair. Front Bioeng Biotechnol 2020; 8:578988. [PMID: 33363124 PMCID: PMC7759629 DOI: 10.3389/fbioe.2020.578988] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Interest is rapidly growing in the design and preparation of bioactive scaffolds, mimicking the biochemical composition and physical microstructure for tissue repair. In this study, a biomimetic biomaterial with nanofibrous architecture composed of silk fibroin and hyaluronic acid (HA) was prepared. Silk fibroin nanofiber was firstly assembled in water and then used as the nanostructural cue; after blending with hyaluronan (silk:HA = 10:1) and the process of freeze-drying, the resulting composite scaffolds exhibited a desirable 3D porous structure and specific nanofiber features. These scaffolds were very porous with the porosity up to 99%. The mean compressive modulus of silk-HA scaffolds with HA MW of 0.6, 1.6, and 2.6 × 106 Da was about 28.3, 30.2, and 29.8 kPa, respectively, all these values were much higher than that of pure silk scaffold (27.5 kPa). This scaffold showed good biocompatibility with bone marrow mesenchymal stem cells, and it enhanced the cellular proliferation significantly when compared with the plain silk fibroin. Collectively, the silk-hyaluronan composite scaffold with a nanofibrous structure and good biocompatibility was successfully prepared, which deserved further exploration as a biomimetic platform for mesenchymal stem cell-based therapy for tissue repair.
Collapse
Affiliation(s)
- Li-Min Yu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Tao Liu
- Department of Textile Engineering, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Yu-Long Ma
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Feng Zhang
- Department of Textile Engineering, College of Textile and Clothing Engineering, Soochow University, Suzhou, China
| | - Yong-Can Huang
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen, China.,Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhi-Hai Fan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
44
|
Mills DK, Luo Y, Elumalai A, Esteve S, Karnik S, Yao S. Creating Structured Hydrogel Microenvironments for Regulating Stem Cell Differentiation. Gels 2020; 6:gels6040047. [PMID: 33276682 PMCID: PMC7768466 DOI: 10.3390/gels6040047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
The development of distinct biomimetic microenvironments for regulating stem cell behavior and bioengineering human tissues and disease models requires a solid understanding of cell-substrate interactions, adhesion, and its role in directing cell behavior, and other physico-chemical cues that drive cell behavior. In the past decade, innovative developments in chemistry, materials science, microfabrication, and associated technologies have given us the ability to manipulate the stem cell microenvironment with greater precision and, further, to monitor effector impacts on stem cells, both spatially and temporally. The influence of biomaterials and the 3D microenvironment's physical and biochemical properties on mesenchymal stem cell proliferation, differentiation, and matrix production are the focus of this review chapter. Mechanisms and materials, principally hydrogel and hydrogel composites for bone and cartilage repair that create "cell-supportive" and "instructive" biomaterials, are emphasized. We begin by providing an overview of stem cells, their unique properties, and their challenges in regenerative medicine. An overview of current fabrication strategies for creating instructive substrates is then reviewed with a focused discussion of selected fabrication methods with an emphasis on bioprinting as a critical tool in creating novel stem cell-based biomaterials. We conclude with a critical assessment of the current state of the field and offer our view on the promises and potential pitfalls of the approaches discussed.
Collapse
Affiliation(s)
- David K. Mills
- School of Biological Sciences, Louisiana Tech University, Ruston, LA 71270, USA;
- Center for Biomedical Engineering and Rehabilitation Science, Louisiana Tech University, Ruston, LA 71270, USA;
- Correspondence:
| | - Yangyang Luo
- Molecular Sciences and Nanotechnology, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Anusha Elumalai
- School of Biological Sciences, Louisiana Tech University, Ruston, LA 71270, USA;
- Center for Biomedical Engineering and Rehabilitation Science, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Savannah Esteve
- Center for Biomedical Engineering and Rehabilitation Science, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, IUPUI, Indianapolis, IN 46202, USA;
| | - Shaomian Yao
- Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803, USA;
| |
Collapse
|
45
|
Park JK, Pham-Nguyen OV, Yoo HS. Coaxial Electrospun Nanofibers with Different Shell Contents to Control Cell Adhesion and Viability. ACS OMEGA 2020; 5:28178-28185. [PMID: 33163800 PMCID: PMC7643203 DOI: 10.1021/acsomega.0c03902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/09/2020] [Indexed: 05/04/2023]
Abstract
Electrospun nanofibers are widely employed as cell culture matrices because their biomimetic structures resemble a natural extracellular matrix. However, due to the limited cell infiltration into nanofibers, three-dimensional (3D) construction of a cell matrix is not easily accomplished. In this study, we developed a method for the partial digestion of a nanofiber into fragmented nanofibers composed of gelatin and polycaprolactone (PCL). The PCL shells of the coaxial fragments were subsequently removed with different concentrations of chloroform to control the remaining PCL on the shell. The swelling and exposure of the gelatin core were manipulated by the remaining PCL shells. When cells were cultivated with the fragmented nanofibers, they were spontaneously assembled on the cell sheets. The cell adhesion and proliferation were significantly affected by the amount of PCL shells on the fragmented nanofibers.
Collapse
Affiliation(s)
- Jae Keun Park
- Department
of Biomedical Materials Engineering, Kangwon
National University, Chuncheon 24341, Republic of Korea
| | - Oanh-Vu Pham-Nguyen
- Department
of Biomedical Materials Engineering, Kangwon
National University, Chuncheon 24341, Republic of Korea
| | - Hyuk Sang Yoo
- Department
of Biomedical Materials Engineering, Kangwon
National University, Chuncheon 24341, Republic of Korea
- Institute
of Bioscience and Biotechnology, Kangwon
National University, Chuncheon 24341, Republic of Korea
- . Website: http://nano-bio.kangwon.ac.kr
| |
Collapse
|
46
|
Kumar P, Saini M, Dehiya BS, Sindhu A, Kumar V, Kumar R, Lamberti L, Pruncu CI, Thakur R. Comprehensive Survey on Nanobiomaterials for Bone Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2019. [PMID: 33066127 PMCID: PMC7601994 DOI: 10.3390/nano10102019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
One of the most important ideas ever produced by the application of materials science to the medical field is the notion of biomaterials. The nanostructured biomaterials play a crucial role in the development of new treatment strategies including not only the replacement of tissues and organs, but also repair and regeneration. They are designed to interact with damaged or injured tissues to induce regeneration, or as a forest for the production of laboratory tissues, so they must be micro-environmentally sensitive. The existing materials have many limitations, including impaired cell attachment, proliferation, and toxicity. Nanotechnology may open new avenues to bone tissue engineering by forming new assemblies similar in size and shape to the existing hierarchical bone structure. Organic and inorganic nanobiomaterials are increasingly used for bone tissue engineering applications because they may allow to overcome some of the current restrictions entailed by bone regeneration methods. This review covers the applications of different organic and inorganic nanobiomaterials in the field of hard tissue engineering.
Collapse
Affiliation(s)
- Pawan Kumar
- Department of Materials Science and Nanotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India; (M.S.); (B.S.D.)
| | - Meenu Saini
- Department of Materials Science and Nanotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India; (M.S.); (B.S.D.)
| | - Brijnandan S. Dehiya
- Department of Materials Science and Nanotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India; (M.S.); (B.S.D.)
| | - Anil Sindhu
- Department of Biotechnology, Deenbandhu Chhotu Ram University of Science and Technology, Murthal 131039, India;
| | - Vinod Kumar
- Department of Bio and Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, India; (V.K.); (R.T.)
| | - Ravinder Kumar
- School of Mechanical Engineering, Lovely Professional University, Phagwara 144411, India
| | - Luciano Lamberti
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, 70125 Bari, Italy;
| | - Catalin I. Pruncu
- Department of Design, Manufacturing & Engineering Management, University of Strathclyde, Glasgow G1 1XJ, UK
- Department of Mechanical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Rajesh Thakur
- Department of Bio and Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, India; (V.K.); (R.T.)
| |
Collapse
|
47
|
Meng QS, Liu J, Wei L, Fan HM, Zhou XH, Liang XT. Senescent mesenchymal stem/stromal cells and restoring their cellular functions. World J Stem Cells 2020; 12:966-985. [PMID: 33033558 PMCID: PMC7524698 DOI: 10.4252/wjsc.v12.i9.966] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/23/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have various properties that make them promising candidates for stem cell-based therapies in clinical settings. These include self-renewal, multilineage differentiation, and immunoregulation. However, recent studies have confirmed that aging is a vital factor that limits their function and therapeutic properties as standardized clinical products. Understanding the features of senescence and exploration of cell rejuvenation methods are necessary to develop effective strategies that can overcome the shortage and instability of MSCs. This review will summarize the current knowledge on characteristics and functional changes of aged MSCs. Additionally, it will highlight cell rejuvenation strategies such as molecular regulation, non-coding RNA modifications, and microenvironment controls that may enhance the therapeutic potential of MSCs in clinical settings.
Collapse
Affiliation(s)
- Qing-Shu Meng
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Jing Liu
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Lu Wei
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Hui-Min Fan
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiao-Hui Zhou
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiao-Ting Liang
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| |
Collapse
|
48
|
Weißenberger M, Weißenberger MH, Wagenbrenner M, Heinz T, Reboredo J, Holzapfel BM, Rudert M, Groll J, Evans CH, Steinert AF. Different types of cartilage neotissue fabricated from collagen hydrogels and mesenchymal stromal cells via SOX9, TGFB1 or BMP2 gene transfer. PLoS One 2020; 15:e0237479. [PMID: 32790806 PMCID: PMC7425924 DOI: 10.1371/journal.pone.0237479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/27/2020] [Indexed: 11/18/2022] Open
Abstract
Objective As native cartilage consists of different phenotypical zones, this study aims to fabricate different types of neocartilage constructs from collagen hydrogels and human mesenchymal stromal cells (MSCs) genetically modified to express different chondrogenic factors. Design Human MSCs derived from bone-marrow of osteoarthritis (OA) hips were genetically modified using adenoviral vectors encoding sex-determining region Y-type high-mobility-group-box (SOX) 9, transforming growth factor beta (TGFB) 1 or bone morphogenetic protein (BMP) 2 cDNA, placed in type I collagen hydrogels and maintained in serum-free chondrogenic media for three weeks. Control constructs contained unmodified MSCs or MSCs expressing GFP. The respective constructs were analyzed histologically, immunohistochemically, biochemically, and by qRT-PCR for chondrogenesis and hypertrophy. Results Chondrogenesis in MSCs was consistently and strongly induced in collagen I hydrogels by the transgenes SOX9, TGFB1 and BMP2 as evidenced by positive staining for proteoglycans, chondroitin-4-sulfate (CS4) and collagen (COL) type II, increased levels of glycosaminoglycan (GAG) synthesis, and expression of mRNAs associated with chondrogenesis. The control groups were entirely non-chondrogenic. The levels of hypertrophy, as judged by expression of alkaline phosphatase (ALP) and COL X on both the protein and mRNA levels revealed different stages of hypertrophy within the chondrogenic groups (BMP2>TGFB1>SOX9). Conclusions Different types of neocartilage with varying levels of hypertrophy could be generated from human MSCs in collagen hydrogels by transfer of genes encoding the chondrogenic factors SOX9, TGFB1 and BMP2. This technology may be harnessed for regeneration of specific zones of native cartilage upon damage.
Collapse
Affiliation(s)
- Manuel Weißenberger
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
- * E-mail:
| | - Manuela H. Weißenberger
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Mike Wagenbrenner
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Tizian Heinz
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Jenny Reboredo
- Department of Tissue Engineering and Regenerative Medicine, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Boris M. Holzapfel
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Maximilian Rudert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Christopher H. Evans
- Department of Physical Medicine and Rehabilitation, Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, MN, United States of America
| | - Andre F. Steinert
- Department of Orthopaedic Surgery, König-Ludwig-Haus, Orthopaedic Center for Musculoskeletal Research (OCMR), Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Wu J, Chen Q, Deng C, Xu B, Zhang Z, Yang Y, Lu T. Exquisite design of injectable Hydrogels in Cartilage Repair. Theranostics 2020; 10:9843-9864. [PMID: 32863963 PMCID: PMC7449920 DOI: 10.7150/thno.46450] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023] Open
Abstract
Cartilage damage is still a threat to human beings, yet there is currently no treatment available to fully restore the function of cartilage. Recently, due to their unique structures and properties, injectable hydrogels have been widely studied and have exhibited high potential for applications in therapeutic areas, especially in cartilage repair. In this review, we briefly introduce the properties of cartilage, some articular cartilage injuries, and now available treatment strategies. Afterwards, we propose the functional and fundamental requirements of injectable hydrogels in cartilage tissue engineering, as well as the main advantages of injectable hydrogels as a therapy for cartilage damage, including strong plasticity and excellent biocompatibility. Moreover, we comprehensively summarize the polymers, cells, and bioactive molecules regularly used in the fabrication of injectable hydrogels, with two kinds of gelation, i.e., physical and chemical crosslinking, which ensure the excellent design of injectable hydrogels for cartilage repair. We also include novel hybrid injectable hydrogels combined with nanoparticles. Finally, we conclude with the advances of this clinical application and the challenges of injectable hydrogels used in cartilage repair.
Collapse
Affiliation(s)
- Jiawei Wu
- Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University School of Life Sciences
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Qi Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, China
| | - Baoping Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Zeiyan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Tingli Lu
- Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University School of Life Sciences
| |
Collapse
|
50
|
Xue J, Pisignano D, Xia Y. Maneuvering the Migration and Differentiation of Stem Cells with Electrospun Nanofibers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000735. [PMID: 32775158 PMCID: PMC7404157 DOI: 10.1002/advs.202000735] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/30/2020] [Indexed: 05/21/2023]
Abstract
Electrospun nanofibers have been extensively explored as a class of scaffolding materials for tissue regeneration, because of their unique capability to mimic some features and functions of the extracellular matrix, including the fibrous morphology and mechanical properties, and to a certain extent the chemical/biological cues. This work reviews recent progress in applying electrospun nanofibers to direct the migration of stem cells and control their differentiation into specific phenotypes. First, the physicochemical properties that make electrospun nanofibers well-suited as a supporting material to expand stem cells by controlling their migration and differentiation are introduced. Then various systems are analyzed in conjunction with mesenchymal, neuronal, and embryonic stem cells, as well as induced pluripotent stem cells. Finally, some perspectives on the challenges and future opportunities in combining electrospun nanofibers with stem cells are offered to address clinical issues.
Collapse
Affiliation(s)
- Jiajia Xue
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Dario Pisignano
- Dipartimento di FisicaUniversità di PisaLargo B. Pontecorvo 3PisaI‐56127Italy
- NESTIstituto Nanoscienze‐CNRPiazza S. Silvestro 12PisaI‐56127Italy
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- School of Chemistry and BiochemistrySchool of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|