51
|
Hu X, Xia Z, Cai K. Recent advances of 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B 2022; 10:1486-1507. [DOI: 10.1039/d1tb02537f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly recognized as resources for disease treatments and regenerative medicine. Meanwhile, the unique chemical and physical properties of hydrogels provide innate advantages to achieve...
Collapse
|
52
|
Abstract
Tissue engineering or tissue reconstruction/repair/regeneration may be considered as a guiding strategy in oral and maxillofacial surgery, as well as in endodontics, orthodontics, periodontics, and daily clinical practice. A wide range of techniques has been developed over the past years, from tissue grafts to the more recent and innovative regenerative procedures. Continuous research in the field of natural and artificial materials and biomaterials, as well as in advanced scaffold design strategies has been carried out. The focus has also been on various growth factors involved in dental tissue repair or reconstruction. Benefiting from the recent literature, this review paper illustrates current innovative strategies and technological approaches in oral and maxillofacial tissue engineering, trying to offer some information regarding the available scientific data and practical applications. After introducing tissue engineering aspects, an overview on additive manufacturing technologies will be provided, with a focus on the applications of superparamagnetic iron oxide nanoparticles in the biomedical field. The potential applications of magnetic fields and magnetic devices on the acceleration of orthodontic tooth movement will be analysed.
Collapse
|
53
|
Kim SH, Kim D, Cha M, Kim SH, Jung Y. The Regeneration of Large-Sized and Vascularized Adipose Tissue Using a Tailored Elastic Scaffold and dECM Hydrogels. Int J Mol Sci 2021; 22:ijms222212560. [PMID: 34830444 PMCID: PMC8624932 DOI: 10.3390/ijms222212560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/03/2022] Open
Abstract
A dome-shaped elastic poly(l-lactide-co-caprolactone) (PLCL) scaffold with a channel and pore structure was fabricated by a combinative method of 3D printing technology and the gel pressing method (13 mm in diameter and 6.5 mm in thickness) for patient-specific regeneration. The PLCL scaffold was combined with adipose decellularized extracellular matrix (adECM) and heart decellularized extracellular matrix (hdECM) hydrogels and human adipose-derived stem cells (hADSCs) to promote adipogenesis and angiogenesis. These scaffolds had mechanical properties similar to those of native adipose tissue for improved tissue regeneration. The results of the in vitro real-time PCR showed that the dECM hydrogel mixture induces adipogenesis. In addition, the in vivo study at 12 weeks demonstrated that the tissue-engineered PLCL scaffolds containing the hydrogel mixture (hdECM/adECM (80:20)) and hADSCs promoted angiogenesis and adipose tissue formation, and suppressed apoptosis. Therefore, we expect that our constructs will be clinically applicable as material for the regeneration of patient-specific large-sized adipose tissue.
Collapse
Affiliation(s)
- Su Hee Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- R&D Center, Medifab Co., Ltd., 70 Dusan-ro, Geumcheon-gu, Seoul 08584, Korea;
| | - Donghak Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
| | - Misun Cha
- R&D Center, Medifab Co., Ltd., 70 Dusan-ro, Geumcheon-gu, Seoul 08584, Korea;
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Korea
- Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
- Correspondence: (S.H.K.); (Y.J.)
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea; (S.H.K.); (D.K.)
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul 03722, Korea
- Correspondence: (S.H.K.); (Y.J.)
| |
Collapse
|
54
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
55
|
Nguyen TPT, Li F, Shrestha S, Tuan RS, Thissen H, Forsythe JS, Frith JE. Cell-laden injectable microgels: Current status and future prospects for cartilage regeneration. Biomaterials 2021; 279:121214. [PMID: 34736147 DOI: 10.1016/j.biomaterials.2021.121214] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/19/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022]
Abstract
Injectable hydrogels have been employed extensively as versatile materials for cartilage regeneration due to their excellent biocompatibility, tunable structure, and ability to accommodate bioactive factors, as well as their ability to be locally delivered via minimally invasive injection to fill irregular defects. More recently, in vitro and in vivo studies have revealed that processing these materials to produce cell-laden microgels can enhance cell-cell and cell-matrix interactions and boost nutrient and metabolite exchange. Moreover, these studies have demonstrated gene expression profiles and matrix regeneration that are superior compared to conventional injectable bulk hydrogels. As cell-laden microgels and their application in cartilage repair are moving closer to clinical translation, this review aims to present an overview of the recent developments in this field. Here we focus on the currently used biomaterials and crosslinking strategies, the innovative fabrication techniques being used for the production of microgels, the cell sources used, the signals used for induction of chondrogenic differentiation and the resultant biological responses, and the ability to create three-dimensional, functional cartilage tissues. In addition, this review also covers the current clinical approaches for repairing cartilage as well as specific challenges faced when attempting the regeneration of damaged cartilage tissue. New findings related to the macroporous nature of the structures formed by the assembled microgel building blocks and the novel use of microgels in 3D printing for cartilage tissue engineering are also highlighted. Finally, we outline the challenges and future opportunities for employing cell-laden microgels in clinical applications.
Collapse
Affiliation(s)
- Thuy P T Nguyen
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Surakshya Shrestha
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Helmut Thissen
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC, 3168, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| | - Jessica E Frith
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, VIC, 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Clayton, VIC 3800, Australia.
| |
Collapse
|
56
|
Tarassoli SP, Jessop ZM, Jovic T, Hawkins K, Whitaker IS. Candidate Bioinks for Extrusion 3D Bioprinting-A Systematic Review of the Literature. Front Bioeng Biotechnol 2021; 9:616753. [PMID: 34722473 PMCID: PMC8548422 DOI: 10.3389/fbioe.2021.616753] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/19/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: Bioprinting is becoming an increasingly popular platform technology for engineering a variety of tissue types. Our aim was to identify biomaterials that have been found to be suitable for extrusion 3D bioprinting, outline their biomechanical properties and biocompatibility towards their application for bioprinting specific tissue types. This systematic review provides an in-depth overview of current biomaterials suitable for extrusion to aid bioink selection for specific research purposes and facilitate design of novel tailored bioinks. Methods: A systematic search was performed on EMBASE, PubMed, Scopus and Web of Science databases according to the PRISMA guidelines. References of relevant articles, between December 2006 to January 2018, on candidate bioinks used in extrusion 3D bioprinting were reviewed by two independent investigators against standardised inclusion and exclusion criteria. Data was extracted on bioprinter brand and model, printing technique and specifications (speed and resolution), bioink material and class of mechanical assessment, cell type, viability, and target tissue. Also noted were authors, study design (in vitro/in vivo), study duration and year of publication. Results: A total of 9,720 studies were identified, 123 of which met inclusion criteria, consisting of a total of 58 reports using natural biomaterials, 26 using synthetic biomaterials and 39 using a combination of biomaterials as bioinks. Alginate (n = 50) and PCL (n = 33) were the most commonly used bioinks, followed by gelatin (n = 18) and methacrylated gelatin (GelMA) (n = 16). Pneumatic extrusion bioprinting techniques were the most common (n = 78), followed by piston (n = 28). The majority of studies focus on the target tissue, most commonly bone and cartilage, and investigate only one bioink rather than assessing a range to identify those with the most promising printability and biocompatibility characteristics. The Bioscaffolder (GeSiM, Germany), 3D Discovery (regenHU, Switzerland), and Bioplotter (EnvisionTEC, Germany) were the most commonly used commercial bioprinters (n = 35 in total), but groups most often opted to create their own in-house devices (n = 20). Many studies also failed to specify whether the mechanical data reflected pre-, during or post-printing, pre- or post-crosslinking and with or without cells. Conclusions: Despite the continued increase in the variety of biocompatible synthetic materials available, there has been a shift change towards using natural rather than synthetic bioinks for extrusion bioprinting, dominated by alginate either alone or in combination with other biomaterials. On qualitative analysis, no link was demonstrated between the type of bioink or extrusion technique and the target tissue, indicating that bioprinting research is in its infancy with no established tissue specific bioinks or bioprinting techniques. Further research is needed on side-by-side characterisation of bioinks with standardisation of the type and timing of biomechanical assessment.
Collapse
Affiliation(s)
- Sam P Tarassoli
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom
| | - Zita M Jessop
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| | - Thomas Jovic
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| | - Karl Hawkins
- Centre for NanoHealth, Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom
| | - Iain S Whitaker
- Reconstructive Surgery & Regenerative Medicine Research Group (ReconRegen), Swansea University Medical School, Institute of Life Sciences, Swansea, United Kingdom.,The Welsh Centre for Burns & Plastic Surgery, Morriston Hospital, Swansea, United Kingdom
| |
Collapse
|
57
|
Sun Y, Chi X, Meng H, Ma M, Wang J, Feng Z, Quan Q, Liu G, Wang Y, Xie Y, Zheng Y, Peng J. Polylysine-decorated macroporous microcarriers laden with adipose-derived stem cells promote nerve regeneration in vivo. Bioact Mater 2021; 6:3987-3998. [PMID: 33997488 PMCID: PMC8082165 DOI: 10.1016/j.bioactmat.2021.03.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cell transplantation is an effective strategy to improve the repair effect of nerve guide conduits (NGCs). However, problems such as low loading efficiency and cell anoikis undermine the outcomes. Microcarriers are efficient 3D cell culture scaffolds, which can also prevent cell anoikis by providing substrate for adhesion during transplantation. Here, we demonstrate for the first time microcarrier-based cell transplantation in peripheral nerve repair. We first prepared macroporous chitosan microcarriers (CSMCs) by the emulsion-phase separation method, and then decorated the CSMCs with polylysine (pl-CSMCs) to improve cell affinity. We then loaded the pl-CSMCs with adipose-derived stem cells (ADSCs) and injected them into electrospun polycaprolactone/chitosan NGCs to repair rat sciatic nerve defects. The ADSCs-laden pl-CSMCs effectively improved nerve regeneration as demonstrated by evaluation of histology, motor function recovery, electrophysiology, and gastrocnemius recovery. With efficient cell transplantation, convenient operation, and the multiple merits of ADSCs, the ADSCs-laden pl-CSMCs hold good potential in peripheral nerve repair.
Collapse
Affiliation(s)
- Yi Sun
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China
| | - Xiaoqi Chi
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Haoye Meng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, PR China
| | - Mengjiao Ma
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Jing Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, PR China
| | - Zhaoxuan Feng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Qi Quan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, PR China
| | - Guodong Liu
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Yansen Wang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Yajie Xie
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Yudong Zheng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, PR China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, PR China
| |
Collapse
|
58
|
Dubay R, Urban JN, Darling EM. Single-Cell Microgels for Diagnostics and Therapeutics. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2009946. [PMID: 36329867 PMCID: PMC9629779 DOI: 10.1002/adfm.202009946] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Indexed: 05/14/2023]
Abstract
Cell encapsulation within hydrogel droplets is transforming what is feasible in multiple fields of biomedical science such as tissue engineering and regenerative medicine, in vitro modeling, and cell-based therapies. Recent advances have allowed researchers to miniaturize material encapsulation complexes down to single-cell scales, where each complex, termed a single-cell microgel, contains only one cell surrounded by a hydrogel matrix while remaining <100 μm in size. With this achievement, studies requiring single-cell resolution are now possible, similar to those done using liquid droplet encapsulation. Of particular note, applications involving long-term in vitro cultures, modular bioinks, high-throughput screenings, and formation of 3D cellular microenvironments can be tuned independently to suit the needs of individual cells and experimental goals. In this progress report, an overview of established materials and techniques used to fabricate single-cell microgels, as well as insight into potential alternatives is provided. This focused review is concluded by discussing applications that have already benefited from single-cell microgel technologies, as well as prospective applications on the cusp of achieving important new capabilities.
Collapse
Affiliation(s)
- Ryan Dubay
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
- Draper, 555 Technology Sq., Cambridge, MA 02139, USA
| | - Joseph N Urban
- Center for Biomedical Engineering, Brown University, 175 Meeting St., Providence, RI 02912, USA
| | - Eric M Darling
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Center for Biomedical Engineering, School of Engineering, Department of Orthopaedics, Brown University, 175 Meeting St., Providence, RI 02912, USA
| |
Collapse
|
59
|
Rahimnejad M, Rezvaninejad R, Rezvaninejad R, França R. Biomaterials in bone and mineralized tissue engineering using 3D printing and bioprinting technologies. Biomed Phys Eng Express 2021; 7. [PMID: 34438382 DOI: 10.1088/2057-1976/ac21ab] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022]
Abstract
This review focuses on recently developed printable biomaterials for bone and mineralized tissue engineering. 3D printing or bioprinting is an advanced technology to design and fabricate complex functional 3D scaffolds, mimicking native tissue forin vivoapplications. We categorized the biomaterials into two main classes: 3D printing and bioprinting. Various biomaterials, including natural, synthetic biopolymers and their composites, have been studied. Biomaterial inks or bioinks used for bone and mineralized tissue regeneration include hydrogels loaded with minerals or bioceramics, cells, and growth factors. In 3D printing, the scaffold is created by acellular biomaterials (biomaterial inks), while in 3D bioprinting, cell-laden hydrogels (bioinks) are used. Two main classes of bioceramics, including bioactive and bioinert ceramics, are reviewed. Bioceramics incorporation provides osteoconductive properties and induces bone formation. Each biopolymer and mineral have its advantages and limitations. Each component of these composite biomaterials provides specific properties, and their combination can ameliorate the mechanical properties, bioactivity, or biological integration of the 3D printed scaffold. Present challenges and future approaches to address them are also discussed.
Collapse
Affiliation(s)
- Maedeh Rahimnejad
- Biomedical Engineering Institute, Université de Montreal, Montreal, QC, Canada
| | - Raziyehsadat Rezvaninejad
- Department of Oral Medicine, Faculty of Dentistry, Hormozgan University of Medical Sciences, Hormozgan, Iran
| | | | - Rodrigo França
- Department of Restorative Dentistry, College of Dentistry, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
60
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Overcoming the Dependence on Animal Models for Osteoarthritis Therapeutics - The Promises and Prospects of In Vitro Models. Adv Healthc Mater 2021; 10:e2100961. [PMID: 34302436 DOI: 10.1002/adhm.202100961] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis (OA) is a musculoskeletal disease characterized by progressive degeneration of osteochondral tissues. Current treatment is restricted to the reduction of pain and loss of function of the joint. To better comprehend the OA pathophysiological conditions, several models are employed, however; there is no consensus on a suitable model. In this review, different in vitro models being developed for possible therapeutic intervention of OA are outlined. Herein, various in vitro OA models starting from 2D model, co-culture model, 3D models, dynamic culture model to advanced technologies-based models such as 3D bioprinting, bioassembly, organoids, and organ-on-chip-based models are discussed with their advantages and disadvantages. Besides, different growth factors, cytokines, and chemicals being utilized for induction of OA condition are reviewed in detail. Furthermore, there is focus on scrutinizing different molecular and possible therapeutic targets for better understanding the mechanisms and OA therapeutics. Finally, the underlying challenges associated with in vitro models are discussed followed by future prospective. Taken together, a comprehensive overview of in vitro OA models, factors to induce OA-like conditions, and intricate molecular targets with the potential to develop personalized osteoarthritis therapeutics in the future with clinical translation is provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Joseph Christakiran Moses
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Nandana Bhardwaj
- Department of Science and Mathematics Indian Institute of Information Technology Guwahati Bongora Guwahati Assam 781015 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
61
|
Rubí-Sans G, Nyga A, Rebollo E, Pérez-Amodio S, Otero J, Navajas D, Mateos-Timoneda MA, Engel E. Development of Cell-Derived Matrices for Three-Dimensional In Vitro Cancer Cell Models. ACS APPLIED MATERIALS & INTERFACES 2021; 13:44108-44123. [PMID: 34494824 DOI: 10.1021/acsami.1c13630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Most morphogenetic and pathological processes are driven by cells responding to the surrounding matrix, such as its composition, architecture, and mechanical properties. Despite increasing evidence for the role of extracellular matrix (ECM) in tissue and disease development, many in vitro substitutes still fail to effectively mimic the native microenvironment. We established a novel method to produce macroscale (>1 cm) mesenchymal cell-derived matrices (CDMs) aimed to mimic the fibrotic tumor microenvironment surrounding epithelial cancer cells. CDMs are produced by human adipose mesenchymal stem cells cultured in sacrificial 3D scaffold templates of fibronectin-coated poly-lactic acid microcarriers (MCs) in the presence of macromolecular crowders. We showed that decellularized CDMs closely mimic the fibrillar protein composition, architecture, and mechanical properties of human fibrotic ECM from cancer masses. CDMs had highly reproducible composition made of collagen types I and III and fibronectin ECM with tunable mechanical properties. Moreover, decellularized and MC-free CDMs were successfully repopulated with cancer cells throughout their 3D structure, and following chemotherapeutic treatment, cancer cells showed greater doxorubicin resistance compared to 3D culture in collagen hydrogels. Collectively, these results support the use of CDMs as a reproducible and tunable tool for developing 3D in vitro cancer models.
Collapse
Affiliation(s)
- Gerard Rubí-Sans
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Agata Nyga
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Soledad Pérez-Amodio
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| | - Jorge Otero
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Spain
- CIBER de Enfermedades Respiratorias, Madrid 28029, Spain
| | - Daniel Navajas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Spain
- CIBER de Enfermedades Respiratorias, Madrid 28029, Spain
| | - Miguel A Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès (Barcelona) 08195, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| |
Collapse
|
62
|
Collagen Bioinks for Bioprinting: A Systematic Review of Hydrogel Properties, Bioprinting Parameters, Protocols, and Bioprinted Structure Characteristics. Biomedicines 2021; 9:biomedicines9091137. [PMID: 34572322 PMCID: PMC8468019 DOI: 10.3390/biomedicines9091137] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/05/2021] [Accepted: 08/27/2021] [Indexed: 01/01/2023] Open
Abstract
Bioprinting is a modern tool suitable for creating cell scaffolds and tissue or organ carriers from polymers that mimic tissue properties and create a natural environment for cell development. A wide range of polymers, both natural and synthetic, are used, including extracellular matrix and collagen-based polymers. Bioprinting technologies, based on syringe deposition or laser technologies, are optimal tools for creating precise constructs precisely from the combination of collagen hydrogel and cells. This review describes the different stages of bioprinting, from the extraction of collagen hydrogels and bioink preparation, over the parameters of the printing itself, to the final testing of the constructs. This study mainly focuses on the use of physically crosslinked high-concentrated collagen hydrogels, which represents the optimal way to create a biocompatible 3D construct with sufficient stiffness. The cell viability in these gels is mainly influenced by the composition of the bioink and the parameters of the bioprinting process itself (temperature, pressure, cell density, etc.). In addition, a detailed table is included that lists the bioprinting parameters and composition of custom bioinks from current studies focusing on printing collagen gels without the addition of other polymers. Last but not least, our work also tries to refute the often-mentioned fact that highly concentrated collagen hydrogel is not suitable for 3D bioprinting and cell growth and development.
Collapse
|
63
|
Drzeniek NM, Mazzocchi A, Schlickeiser S, Forsythe SD, Moll G, Geißler S, Reinke P, Gossen M, Gorantla VS, Volk HD, Soker S. Bio-instructive hydrogel expands the paracrine potency of mesenchymal stem cells. Biofabrication 2021; 13:10.1088/1758-5090/ac0a32. [PMID: 34111862 PMCID: PMC10024818 DOI: 10.1088/1758-5090/ac0a32] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/10/2021] [Indexed: 02/03/2023]
Abstract
The therapeutic efficacy of clinically applied mesenchymal stromal cells (MSCs) is limited due to their injection into harshin vivoenvironments, resulting in the significant loss of their secretory function upon transplantation. A potential strategy for preserving their full therapeutic potential is encapsulation of MSCs in a specialized protective microenvironment, for example hydrogels. However, commonly used injectable hydrogels for cell delivery fail to provide the bio-instructive cues needed to sustain and stimulate cellular therapeutic functions. Here we introduce a customizable collagen I-hyaluronic acid (COL-HA)-based hydrogel platform for the encapsulation of MSCs. Cells encapsulated within COL-HA showed a significant expansion of their secretory profile compared to MSCs cultured in standard (2D) cell culture dishes or encapsulated in other hydrogels. Functionalization of the COL-HA backbone with thiol-modified glycoproteins such as laminin led to further changes in the paracrine profile of MSCs. In depth profiling of more than 250 proteins revealed an expanded secretion profile of proangiogenic, neuroprotective and immunomodulatory paracrine factors in COL-HA-encapsulated MSCs with a predicted augmented pro-angiogenic potential. This was confirmed by increased capillary network formation of endothelial cells stimulated by conditioned media from COL-HA-encapsulated MSCs. Our findings suggest that encapsulation of therapeutic cells in a protective COL-HA hydrogel layer provides the necessary bio-instructive cues to maintain and direct their therapeutic potential. Our customizable hydrogel combines bioactivity and clinically applicable properties such as injectability, on-demand polymerization and tissue-specific elasticity, all features that will support and improve the ability to successfully deliver functional MSCs into patients.
Collapse
Affiliation(s)
- Norman M Drzeniek
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andrea Mazzocchi
- Known Medicine Inc., 675 Arapeen Dr, Suite 103A-1, Salt Lake City, UT 84108, United States of America.,Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Stephan Schlickeiser
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Steven D Forsythe
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Guido Moll
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sven Geißler
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Manfred Gossen
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin 13353, Germany.,Institute of Active Polymers, Helmholtz-Zentrum Hereon, Kantstr. 55, Teltow 14513, Germany
| | - Vijay S Gorantla
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Hans-Dieter Volk
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States of America
| |
Collapse
|
64
|
Wang Z, Wang L, Li T, Liu S, Guo B, Huang W, Wu Y. 3D bioprinting in cardiac tissue engineering. Am J Cancer Res 2021; 11:7948-7969. [PMID: 34335973 PMCID: PMC8315053 DOI: 10.7150/thno.61621] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/06/2021] [Indexed: 12/22/2022] Open
Abstract
Heart disease is the main cause of death worldwide. Because death of the myocardium is irreversible, it remains a significant clinical challenge to rescue myocardial deficiency. Cardiac tissue engineering (CTE) is a promising strategy for repairing heart defects and offers platforms for studying cardiac tissue. Numerous achievements have been made in CTE in the past decades based on various advanced engineering approaches. 3D bioprinting has attracted much attention due to its ability to integrate multiple cells within printed scaffolds with complex 3D structures, and many advancements in bioprinted CTE have been reported recently. Herein, we review the recent progress in 3D bioprinting for CTE. After a brief overview of CTE with conventional methods, the current 3D printing strategies are discussed. Bioink formulations based on various biomaterials are introduced, and strategies utilizing composite bioinks are further discussed. Moreover, several applications including heart patches, tissue-engineered cardiac muscle, and other bionic structures created via 3D bioprinting are summarized. Finally, we discuss several crucial challenges and present our perspective on 3D bioprinting techniques in the field of CTE.
Collapse
|
65
|
Yin S, Cao Y. Hydrogels for Large-Scale Expansion of Stem Cells. Acta Biomater 2021; 128:1-20. [PMID: 33746032 DOI: 10.1016/j.actbio.2021.03.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summarize current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.
Collapse
Affiliation(s)
- Sheng Yin
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057.
| |
Collapse
|
66
|
Non-mulberry silk fiber-based scaffolds reinforced by PLLA porous microspheres for auricular cartilage: An in vitro study. Int J Biol Macromol 2021; 182:1704-1712. [PMID: 34052269 DOI: 10.1016/j.ijbiomac.2021.05.145] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022]
Abstract
Designing clinical applicable polymeric composite scaffolds for auricular cartilage tissue engineering requires appropriate mechanical strength and biological characteristics. In this study, silk fiber-based scaffolds co-reinforced with poly-L-lactic acid porous microspheres (PLLA PMs) combined with either Bombyx mori (Bm) or Antheraea pernyi (Ap) silk fibers were fabricated as inspired by the "steel bars reinforced concrete" structure in architecture and their chondrogenic functions were also investigated. We found that the Ap silk fiber-based scaffolds reinforced by PLLA PMs (MAF) exhibited superior physical properties (the mechanical properties in particular) as compared to the Bm silk fiber-based scaffolds reinforced by PLLA PMs (MBF). Furthermore, in vitro evaluation of chondrogenic potential showed that the MAF provided better cell adhesion, viability, proliferation and GAG secretion than the MBF. Therefore, the MAF are promising in auricular cartilage tissue engineering and relevant plastic surgery-related applications.
Collapse
|
67
|
Jiang S, Wang M, He J. A review of biomimetic scaffolds for bone regeneration: Toward a cell-free strategy. Bioeng Transl Med 2021; 6:e10206. [PMID: 34027093 PMCID: PMC8126827 DOI: 10.1002/btm2.10206] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
In clinical terms, bone grafting currently involves the application of autogenous, allogeneic, or xenogeneic bone grafts, as well as natural or artificially synthesized materials, such as polymers, bioceramics, and other composites. Many of these are associated with limitations. The ideal scaffold for bone tissue engineering should provide mechanical support while promoting osteogenesis, osteoconduction, and even osteoinduction. There are various structural complications and engineering difficulties to be considered. Here, we describe the biomimetic possibilities of the modification of natural or synthetic materials through physical and chemical design to facilitate bone tissue repair. This review summarizes recent progresses in the strategies for constructing biomimetic scaffolds, including ion-functionalized scaffolds, decellularized extracellular matrix scaffolds, and micro- and nano-scale biomimetic scaffold structures, as well as reactive scaffolds induced by physical factors, and other acellular scaffolds. The fabrication techniques for these scaffolds, along with current strategies in clinical bone repair, are described. The developments in each category are discussed in terms of the connection between the scaffold materials and tissue repair, as well as the interactions with endogenous cells. As the advances in bone tissue engineering move toward application in the clinical setting, the demonstration of the therapeutic efficacy of these novel scaffold designs is critical.
Collapse
Affiliation(s)
- Sijing Jiang
- Department of Plastic SurgeryFirst Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiChina
| | - Mohan Wang
- Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceHefeiChina
| | - Jiacai He
- Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceHefeiChina
| |
Collapse
|
68
|
Zhang J, Wehrle E, Rubert M, Müller R. 3D Bioprinting of Human Tissues: Biofabrication, Bioinks, and Bioreactors. Int J Mol Sci 2021; 22:ijms22083971. [PMID: 33921417 PMCID: PMC8069718 DOI: 10.3390/ijms22083971] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The field of tissue engineering has progressed tremendously over the past few decades in its ability to fabricate functional tissue substitutes for regenerative medicine and pharmaceutical research. Conventional scaffold-based approaches are limited in their capacity to produce constructs with the functionality and complexity of native tissue. Three-dimensional (3D) bioprinting offers exciting prospects for scaffolds fabrication, as it allows precise placement of cells, biochemical factors, and biomaterials in a layer-by-layer process. Compared with traditional scaffold fabrication approaches, 3D bioprinting is better to mimic the complex microstructures of biological tissues and accurately control the distribution of cells. Here, we describe recent technological advances in bio-fabrication focusing on 3D bioprinting processes for tissue engineering from data processing to bioprinting, mainly inkjet, laser, and extrusion-based technique. We then review the associated bioink formulation for 3D bioprinting of human tissues, including biomaterials, cells, and growth factors selection. The key bioink properties for successful bioprinting of human tissue were summarized. After bioprinting, the cells are generally devoid of any exposure to fluid mechanical cues, such as fluid shear stress, tension, and compression, which are crucial for tissue development and function in health and disease. The bioreactor can serve as a simulator to aid in the development of engineering human tissues from in vitro maturation of 3D cell-laden scaffolds. We then describe some of the most common bioreactors found in the engineering of several functional tissues, such as bone, cartilage, and cardiovascular applications. In the end, we conclude with a brief insight into present limitations and future developments on the application of 3D bioprinting and bioreactor systems for engineering human tissue.
Collapse
|
69
|
Decante G, Costa JB, Silva-Correia J, Collins MN, Reis RL, Oliveira JM. Engineering bioinks for 3D bioprinting. Biofabrication 2021; 13. [PMID: 33662949 DOI: 10.1088/1758-5090/abec2c] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
In recent years, three-dimensional (3D) bioprinting has attracted wide research interest in biomedical engineering and clinical applications. This technology allows for unparalleled architecture control, adaptability and repeatability that can overcome the limits of conventional biofabrication techniques. Along with the emergence of a variety of 3D bioprinting methods, bioinks have also come a long way. From their first developments to support bioprinting requirements, they are now engineered to specific injury sites requirements to mimic native tissue characteristics and to support biofunctionality. Current strategies involve the use of bioinks loaded with cells and biomolecules of interest, without altering their functions, to deliverin situthe elements required to enhance healing/regeneration. The current research and trends in bioink development for 3D bioprinting purposes is overviewed herein.
Collapse
Affiliation(s)
- Guy Decante
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João B Costa
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maurice N Collins
- Bernal Institute, School of Engineering, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
70
|
Abstract
Polylactic acid (PLA) is the most widely used raw material in extrusion-based three-dimensional (3D) printing (fused deposition modeling, FDM approach) in many areas since it is biodegradable and environmentally friendly, however its utilization is limited due to some of its disadvantages such as mechanical weakness, water solubility rate, etc. FDM is a simple and more cost-effective fabrication process compared to other 3D printing techniques. Unfortunately, there are deficiencies of the FDM approach, such as mechanical weakness of the FDM parts compared to the parts produced by the conventional injection and compression molding methods. Preparation of PLA composites with suitable additives is the most useful technique to improve the properties of the 3D-printed PLA parts obtained by the FDM method. In the last decade, newly developed PLA composites find large usage areas both in academic and industrial circles. This review focuses on the chemistry and properties of pure PLA and also the preparation methods of the PLA composites which will be used as a raw material in 3D printers. The main drawbacks of the pure PLA filaments and the necessity for the preparation of PLA composites which will be employed in the FDM-based 3D printing applications is also discussed in the first part. The current methods to obtain PLA composites as raw materials to be used as filaments in the extrusion-based 3D printing are given in the second part. The applications of the novel PLA composites by utilizing the FDM-based 3D printing technology in the fields of biomedical, tissue engineering, human bone repair, antibacterial, bioprinting, electrical conductivity, electromagnetic, sensor, battery, automotive, aviation, four-dimensional (4D) printing, smart textile, environmental, and luminescence applications are presented and critically discussed in the third part of this review.
Collapse
|
71
|
Rubí-Sans G, Cano-Torres I, Pérez-Amodio S, Blanco-Fernandez B, Mateos-Timoneda MA, Engel E. Development and Angiogenic Potential of Cell-Derived Microtissues Using Microcarrier-Template. Biomedicines 2021; 9:232. [PMID: 33669131 PMCID: PMC8025087 DOI: 10.3390/biomedicines9030232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Tissue engineering and regenerative medicine approaches use biomaterials in combination with cells to regenerate lost functions of tissues and organs to prevent organ transplantation. However, most of the current strategies fail in mimicking the tissue's extracellular matrix properties. In order to mimic native tissue conditions, we developed cell-derived matrix (CDM) microtissues (MT). Our methodology uses poly-lactic acid (PLA) and Cultispher® S microcarriers' (MCs') as scaffold templates, which are seeded with rat bone marrow mesenchymal stem cells (rBM-MSCs). The scaffold template allows cells to generate an extracellular matrix, which is then extracted for downstream use. The newly formed CDM provides cells with a complex physical (MT architecture) and biochemical (deposited ECM proteins) environment, also showing spontaneous angiogenic potential. Our results suggest that MTs generated from the combination of these two MCs (mixed MTs) are excellent candidates for tissue vascularization. Overall, this study provides a methodology for in-house fabrication of microtissues with angiogenic potential for downstream use in various tissue regenerative strategies.
Collapse
Affiliation(s)
- Gerard Rubí-Sans
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - Irene Cano-Torres
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - Soledad Pérez-Amodio
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
- IMEM-BRT Group, Department of Material Science, Escola d'Enginyeria de Barcelona Est (EEBE), Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| | - Barbara Blanco-Fernandez
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - Miguel A Mateos-Timoneda
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Bioengineering Institute of Technology, Department of Basic Science, Universitat Internacional de Catalunya (UIC), 08195 Barcelona, Spain
| | - Elisabeth Engel
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
- IMEM-BRT Group, Department of Material Science, Escola d'Enginyeria de Barcelona Est (EEBE), Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| |
Collapse
|
72
|
Xiong X, Xiao W, Zhou S, Cui R, Xu HHK, Qu S. Enhanced proliferation and angiogenic phenotype of endothelial cells via negatively-charged alginate and chondroitin sulfate microsphere hydrogels. Biomed Mater 2021; 16:025012. [PMID: 33412523 DOI: 10.1088/1748-605x/abd994] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Sodium alginate-based hydrogel was the one of the most used polymers for cell delivery. However, the adsorption of extracellular matrix and proteins was inhibited due to the formation of a hydrated surface layer of these hydrogels. In this study, a novel cell delivery system, negatively-charged alginate and chondroitin sulfate microsphere hydrogel (nCACSMH), was fabricated with excellent permeability and biocompatibility in the action of a high voltage direct-current electric field. Negative charge was introduced to the surface of nCACSMH to obtain the expanded network and enhanced permeability. Additionally, the increasing content of chondroitin sulfate in nCACSMH could give rise to the charge density and its asymmetric structure, thus the uneven, plicate and expanded surface of nCACSMH which was favorable to cell proliferation was developed. Moreover, chondroitin sulfate was released with the degradation of nCACSMH, which played a crucial role in maintaining the normal physiological functions of cells. Thus the proliferation of human umbilical vein endothelial cells (HUVECs) was further accelerated and the angiogenesis related genes expression in endothelial cells was continuously and dramatically up-regulated. After 4 d, the proliferation and viability of HUVECs were significantly improved, the cells were distributed evenly in nCACSMH. The novel nCACSMH has the potential to be used as cell delivery, three-dimensional (3D) cell cultures for cell therapy, 3D bioprinting, high-throughput screening for drugs, and disease model for regeneration and constructing of tissue engineering.
Collapse
Affiliation(s)
- Xiong Xiong
- Key Lab of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China. School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China. Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD 21201, United States of America. These authors contributed to this work equally
| | | | | | | | | | | |
Collapse
|
73
|
Deng Z, Jin J, Wang S, Qi F, Chen X, Liu C, Li Y, Ma Y, Lyu F, Zheng Q. Narrative review of the choices of stem cell sources and hydrogels for cartilage tissue engineering. ANNALS OF TRANSLATIONAL MEDICINE 2021; 8:1598. [PMID: 33437797 PMCID: PMC7791208 DOI: 10.21037/atm-20-2342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Stem cell-based therapy is a promising treatment for cartilage defects due to the pluripotency, abundant sources and low immunogenicity of stem cells. Hydrogels are a promising class of biomaterials for cartilage engineering and are characterized by bioactivity, degradability and elasticity as well as provide water content and mechanical support. The combination of stem cells and hydrogels opens new possibilities for cartilage tissue engineering. However, the selection of suitable types of stem cells and hydrogels is difficult. Currently, various types of stem cells, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and peripheral blood mononuclear cells (PBMSCs), and various types of hydrogels, including natural polymers, chemically modified natural polymers and synthetic polymers, have been explored based on their potential for cartilage tissue engineering. These materials are used independently or in combination; however, there is no clear understanding of their merits and disadvantages with regard to their suitability for cartilage repair. In this article, we aim to review recent progress in the use of stem cell-hydrogel hybrid constructs for cartilage tissue engineering. We focus on the effects of stem cell types and hydrogel types on efficient chondrogenesis from cellular, preclinical and clinical perspectives. We compare and analyze the advantages and disadvantages of these cells and hydrogels with the hope of increasing discussion of their suitability for cartilage repair and present our perspective on their use for the improvement of physical and biological properties for cartilage tissue engineering.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jiewen Jin
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuai Wang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fangjie Qi
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xuepan Chen
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chang Liu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanbing Li
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fengjuan Lyu
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,South China University of Technology-the University of Western Australia Joint Center for Regenerative Medicine Research, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
74
|
Bosch-Rué E, Diez-Tercero L, Giordano-Kelhoffer B, Delgado LM, Bosch BM, Hoyos-Nogués M, Mateos-Timoneda MA, Tran PA, Gil FJ, Perez RA. Biological Roles and Delivery Strategies for Ions to Promote Osteogenic Induction. Front Cell Dev Biol 2021; 8:614545. [PMID: 33520992 PMCID: PMC7841204 DOI: 10.3389/fcell.2020.614545] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Bone is the most studied tissue in the field of tissue regeneration. Even though it has intrinsic capability to regenerate upon injury, several pathologies and injuries could hamper the highly orchestrated bone formation and resorption process. Bone tissue engineering seeks to mimic the extracellular matrix of the tissue and the different biochemical pathways that lead to successful regeneration. For many years, the use of extrinsic factors (i.e., growth factors and drugs) to modulate these biological processes have been the preferred choice in the field. Even though it has been successful in some instances, this approach presents several drawbacks, such as safety-concerns, short release profile and half-time life of the compounds. On the other hand, the use of inorganic ions has attracted significant attention due to their therapeutic effects, stability and lower biological risks. Biomaterials play a key role in such strategies where they serve as a substrate for the incorporation and release of the ions. In this review, the methodologies used to incorporate ions in biomaterials is presented, highlighting the osteogenic properties of such ions and the roles of biomaterials in controlling their release.
Collapse
Affiliation(s)
- Elia Bosch-Rué
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Leire Diez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Luis M. Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Begoña M. Bosch
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Mireia Hoyos-Nogués
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Phong A. Tran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Francisco Javier Gil
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Roman A. Perez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Barcelona, Spain
| |
Collapse
|
75
|
Vanaei S, Parizi M, Vanaei S, Salemizadehparizi F, Vanaei H. An Overview on Materials and Techniques in 3D Bioprinting Toward Biomedical Application. ENGINEERED REGENERATION 2021. [DOI: 10.1016/j.engreg.2020.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
76
|
Wu Y, Kennedy P, Bonazza N, Yu Y, Dhawan A, Ozbolat I. Three-Dimensional Bioprinting of Articular Cartilage: A Systematic Review. Cartilage 2021; 12:76-92. [PMID: 30373384 PMCID: PMC7755962 DOI: 10.1177/1947603518809410] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Treatment of chondral injury is clinically challenging. Available chondral repair/regeneration techniques have significant shortcomings. A viable and durable tissue engineering strategy for articular cartilage repair remains an unmet need. Our objective was to systematically evaluate the published data on bioprinted articular cartilage with regards to scaffold-based, scaffold-free and in situ cartilage bioprinting. DESIGN We performed a systematic review of studies using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. PubMed and ScienceDirect databases were searched and all articles evaluating the use of 3-dimensional (3D) bioprinting in articular cartilage were included. Inclusion criteria included studies written in or translated to English, published in a peer-reviewed journal, and specifically discussing bioinks and/or bioprinting of living cells related to articular cartilage applications. Review papers, articles in a foreign language, and studies not involving bioprinting of living cells related to articular cartilage applications were excluded. RESULTS Twenty-seven studies for articular cartilage bioprinting were identified that met inclusion and exclusion criteria. The technologies, materials, cell types used in these studies, and the biological and physical properties of the created constructs have been demonstrated. CONCLUSION These 27 studies have demonstrated 3D bioprinting of articular cartilage to be a tissue engineering strategy that has tremendous potential translational value. The unique abilities of the varied techniques allow replication of mechanical properties and advances toward zonal differentiation. This review demonstrates that bioprinting has great capacity for clinical cartilage reconstruction and future in vivo implantation.
Collapse
Affiliation(s)
- Yang Wu
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA,The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - Patrick Kennedy
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Nicholas Bonazza
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yin Yu
- Institute for Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China,University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Aman Dhawan
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Ibrahim Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA,The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA,Biomedical Engineering Department, Penn State University, University Park, PA, USA,Materials Research Institute, Penn State University, University Park, PA, USA,Ibrahim Tarik Ozbolat, Penn State University, W313 Millennium Science Complex, University Park, PA 16802, USA.
| |
Collapse
|
77
|
Askari M, Afzali Naniz M, Kouhi M, Saberi A, Zolfagharian A, Bodaghi M. Recent progress in extrusion 3D bioprinting of hydrogel biomaterials for tissue regeneration: a comprehensive review with focus on advanced fabrication techniques. Biomater Sci 2021; 9:535-573. [DOI: 10.1039/d0bm00973c] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Over the last decade, 3D bioprinting has received immense attention from research communities to bridge the divergence between artificially engineered tissue constructs and native tissues.
Collapse
Affiliation(s)
- Mohsen Askari
- Department of Engineering
- School of Science and Technology
- Nottingham Trent University
- Nottingham NG11 8NS
- UK
| | - Moqaddaseh Afzali Naniz
- Department of Engineering
- School of Science and Technology
- Nottingham Trent University
- Nottingham NG11 8NS
- UK
| | - Monireh Kouhi
- Biomaterials Research Group
- Department of Materials Engineering
- Isfahan University of Technology
- Isfahan
- Iran
| | - Azadeh Saberi
- Nanotechnology and Advanced Materials Department
- Materials and Energy Research Center
- Tehran
- Iran
| | | | - Mahdi Bodaghi
- Department of Engineering
- School of Science and Technology
- Nottingham Trent University
- Nottingham NG11 8NS
- UK
| |
Collapse
|
78
|
Wang Z, Kapadia W, Li C, Lin F, Pereira RF, Granja PL, Sarmento B, Cui W. Tissue-specific engineering: 3D bioprinting in regenerative medicine. J Control Release 2021; 329:237-256. [DOI: 10.1016/j.jconrel.2020.11.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022]
|
79
|
Ratheesh G, Vaquette C, Xiao Y. Patient-Specific Bone Particles Bioprinting for Bone Tissue Engineering. Adv Healthc Mater 2020; 9:e2001323. [PMID: 33166078 DOI: 10.1002/adhm.202001323] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Although bioinks with both high printability and shape fidelity while maintaining high cell viability are developed, the biofunctionality of the resulting bioprinted construct is often overlooked. To address this, a methacrylated gelatin (GelMA)-based bioink biofunctionalized with bone particles (BPs) is developed as a personalized treatment strategy for bone regeneration. The bioink consists of incorporating BPs of various sizes (0-500 µm) in GelMA at various concentrations (ranging from 5 to 15% w/v). The printability of the bioink is systematically investigated and it is demonstrated that a 15% w/v BP-loading results in high print quality for 10% and 12.5% GelMA concentrations. Rheological evaluation reveals a strong shear thinning behavior essential for printing and a high gel strength in bioink with 15% w/v 0-500 µm BPs for both GelMA concentrations. In addition, the printability of the bioink and the metabolic activity of the resulting scaffolds are dependent on both the concentration of hydrogel and size of the BPs. Importantly, the cells initially contained in the BPs are able to migrate and colonize the bioprinted scaffold while maintaining their capacity to express early osteogenic markers. This study demonstrates the feasibility of bioprinted viable BPs and may have some potential for chairside clinical translation.
Collapse
Affiliation(s)
- Greeshma Ratheesh
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| | - Cedryck Vaquette
- School of Dentistry, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| |
Collapse
|
80
|
Hogan KJ, Mikos AG. Biodegradable thermoresponsive polymers: Applications in drug delivery and tissue engineering. POLYMER 2020. [DOI: 10.1016/j.polymer.2020.123063] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
81
|
Boularaoui S, Al Hussein G, Khan KA, Christoforou N, Stefanini C. An overview of extrusion-based bioprinting with a focus on induced shear stress and its effect on cell viability. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.bprint.2020.e00093] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
82
|
Chandika P, Heo SY, Kim TH, Oh GW, Kim GH, Kim MS, Jung WK. Recent advances in biological macromolecule based tissue-engineered composite scaffolds for cardiac tissue regeneration applications. Int J Biol Macromol 2020; 164:2329-2357. [DOI: 10.1016/j.ijbiomac.2020.08.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
|
83
|
Matichescu A, Ardelean LC, Rusu LC, Craciun D, Bratu EA, Babucea M, Leretter M. Advanced Biomaterials and Techniques for Oral Tissue Engineering and Regeneration-A Review. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E5303. [PMID: 33238625 PMCID: PMC7700200 DOI: 10.3390/ma13225303] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
The reconstruction or repair of oral and maxillofacial functionalities and aesthetics is a priority for patients affected by tooth loss, congenital defects, trauma deformities, or various dental diseases. Therefore, in dental medicine, tissue reconstruction represents a major interest in oral and maxillofacial surgery, periodontics, orthodontics, endodontics, and even daily clinical practice. The current clinical approaches involve a vast array of techniques ranging from the traditional use of tissue grafts to the most innovative regenerative procedures, such as tissue engineering. In recent decades, a wide range of both artificial and natural biomaterials and scaffolds, genes, stem cells isolated from the mouth area (dental follicle, deciduous teeth, periodontal ligament, dental pulp, salivary glands, and adipose tissue), and various growth factors have been tested in tissue engineering approaches in dentistry, with many being proven successful. However, to fully eliminate the problems of traditional bone and tissue reconstruction in dentistry, continuous research is needed. Based on a recent literature review, this paper creates a picture of current innovative strategies applying dental stem cells for tissue regeneration in different dental fields and maxillofacial surgery, and offers detailed information regarding the available scientific data and practical applications.
Collapse
Affiliation(s)
- Anamaria Matichescu
- Department of Preventive Dentistry, Community and Oral Health, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Lavinia Cosmina Ardelean
- Department of Technology of Materials and Devices in Dental Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Laura-Cristina Rusu
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Dragos Craciun
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Emanuel Adrian Bratu
- Department of Implant Supported Restorations, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Marius Babucea
- Department of Oral Pathology, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania; (L.-C.R.); (D.C.); (M.B.)
| | - Marius Leretter
- Department of Prosthodontics, “Victor Babeș” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Sq., 300041 Timisoara, Romania;
| |
Collapse
|
84
|
Chung JJ, Im H, Kim SH, Park JW, Jung Y. Toward Biomimetic Scaffolds for Tissue Engineering: 3D Printing Techniques in Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:586406. [PMID: 33251199 PMCID: PMC7671964 DOI: 10.3389/fbioe.2020.586406] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022] Open
Abstract
Three-dimensional (3D) printing technology allows fabricating complex and precise structures by stacking materials layer by layer. The fabrication method has a strong potential in the regenerative medicine field to produce customizable and defect-fillable scaffolds for tissue regeneration. Plus, biocompatible materials, bioactive molecules, and cells can be printed together or separately to enhance scaffolds, which can save patients who suffer from shortage of transplantable organs. There are various 3D printing techniques that depend on the types of materials, or inks, used. Here, different types of organs (bone, cartilage, heart valve, liver, and skin) that are aided by 3D printed scaffolds and printing methods that are applied in the biomedical fields are reviewed.
Collapse
Affiliation(s)
- Justin J. Chung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
| | - Heejung Im
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
| | - Soo Hyun Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
| | - Jong Woong Park
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, South Korea
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul, South Korea
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, South Korea
| |
Collapse
|
85
|
Vurat MT, Şeker Ş, Lalegül-Ülker Ö, Parmaksiz M, Elçin AE, Elçin YM. Development of a multicellular 3D-bioprinted microtissue model of human periodontal ligament-alveolar bone biointerface: Towards a pre-clinical model of periodontal diseases and personalized periodontal tissue engineering. Genes Dis 2020; 9:1008-1023. [PMID: 35685479 PMCID: PMC9170773 DOI: 10.1016/j.gendis.2020.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/20/2022] Open
Abstract
While periodontal (PD) disease is among principal causes of tooth loss worldwide, regulation of concomitant soft and mineralized PD tissues, and PD pathogenesis have not been completely clarified yet. Besides, relevant pre-clinical models and in vitro platforms have limitations in simulating human physiology. Here, we have harnessed three-dimensional bioprinting (3DBP) technology for developing a multi-cellular microtissue model resembling PD ligament-alveolar bone (PDL-AB) biointerface for the first time. 3DBP parameters were optimized; the physical, chemical, rheological, mechanical, and thermal properties of the constructs were assessed. Constructs containing gelatin methacryloyl (Gel-MA) and hydroxyapatite-magnetic iron oxide nanoparticles showed higher level of compressive strength when compared with that of Gel-MA constructs. Bioprinted self-supporting microtissue was cultured under flow in a microfluidic platform for >10 days without significant loss of shape fidelity. Confocal microscopy analysis indicated that encapsulated cells were homogenously distributed inside the matrix and preserved their viability for >7 days under microfluidic conditions. Immunofluorescence analysis showed the cohesion of stromal cell surface marker-1+ human PDL fibroblasts containing PDL layer with the osteocalcin+ human osteoblasts containing mineralized layer in time, demonstrating some permeability of the printed constructs to cell migration. Preliminary tetracycline interaction study indicated the uptake of model drug by the cells inside the 3D-microtissue. Also, the non-toxic levels of tetracycline were determined for the encapsulated cells. Thus, the effects of tetracyclines on PDL-AB have clinical significance for treating PD diseases. This 3D-bioprinted multi-cellular periodontal/osteoblastic microtissue model has potential as an in vitro platform for studying processes of the human PDL.
Collapse
|
86
|
Kupikowska-Stobba B, Lewińska D. Polymer microcapsules and microbeads as cell carriers for in vivo biomedical applications. Biomater Sci 2020; 8:1536-1574. [PMID: 32110789 DOI: 10.1039/c9bm01337g] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Polymer microcarriers are being extensively explored as cell delivery vehicles in cell-based therapies and hybrid tissue and organ engineering. Spherical microcarriers are of particular interest due to easy fabrication and injectability. They include microbeads, composed of a porous matrix, and microcapsules, where matrix core is additionally covered with a semipermeable membrane. Microcarriers provide cell containment at implantation site and protect the cells from host immunoresponse, degradation and shear stress. Immobilized cells may be genetically altered to release a specific therapeutic product directly at the target site, eliminating side effects of systemic therapies. Cell microcarriers need to fulfil a number of extremely high standards regarding their biocompatibility, cytocompatibility, immunoisolating capacity, transport, mechanical and chemical properties. To obtain cell microcarriers of specified parameters, a wide variety of polymers, both natural and synthetic, and immobilization methods can be applied. Yet so far, only a few approaches based on cell-laden microcarriers have reached clinical trials. The main issue that still impedes progress of these systems towards clinical application is limited cell survival in vivo. Herein, we review polymer biomaterials and methods used for fabrication of cell microcarriers for in vivo biomedical applications. We describe their key limitations and modifications aiming at improvement of microcarrier in vivo performance. We also present the main applications of polymer cell microcarriers in regenerative medicine, pancreatic islet and hepatocyte transplantation and in the treatment of cancer. Lastly, we outline the main challenges in cell microimmobilization for biomedical purposes, the strategies to overcome these issues and potential future improvements in this area.
Collapse
Affiliation(s)
- Barbara Kupikowska-Stobba
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| | - Dorota Lewińska
- Laboratory of Electrostatic Methods of Bioencapsulation, Department of Biomaterials and Biotechnological Systems, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland.
| |
Collapse
|
87
|
Donate R, Monzón M, Alemán-Domínguez ME. Additive manufacturing of PLA-based scaffolds intended for bone regeneration and strategies to improve their biological properties. E-POLYMERS 2020. [DOI: 10.1515/epoly-2020-0046] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractPolylactic acid (PLA) is one of the most commonly used materials in the biomedical sector because of its processability, mechanical properties and biocompatibility. Among the different techniques that are feasible to process this biomaterial, additive manufacturing (AM) has gained attention recently, as it provides the possibility of tuning the design of the structures. This flexibility in the design stage allows the customization of the parts in order to optimize their use in the tissue engineering field. In the recent years, the application of PLA for the manufacture of bone scaffolds has been especially relevant, since numerous studies have proven the potential of this biomaterial for bone regeneration. This review contains a description of the specific requirements in the regeneration of bone and how the state of the art have tried to address them with different strategies to develop PLA-based scaffolds by AM techniques and with improved biofunctionality.
Collapse
Affiliation(s)
- Ricardo Donate
- Departamento de Ingeniería Mecánica, Universidad de Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas, Spain
| | - Mario Monzón
- Departamento de Ingeniería Mecánica, Universidad de Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas, Spain
| | - María Elena Alemán-Domínguez
- Departamento de Ingeniería Mecánica, Universidad de Las Palmas de Gran Canaria, Campus Universitario de Tafira s/n, 35017, Las Palmas, Spain
| |
Collapse
|
88
|
West-Livingston LN, Park J, Lee SJ, Atala A, Yoo JJ. The Role of the Microenvironment in Controlling the Fate of Bioprinted Stem Cells. Chem Rev 2020; 120:11056-11092. [PMID: 32558555 PMCID: PMC7676498 DOI: 10.1021/acs.chemrev.0c00126] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of tissue engineering and regenerative medicine has made numerous advances in recent years in the arena of fabricating multifunctional, three-dimensional (3D) tissue constructs. This can be attributed to novel approaches in the bioprinting of stem cells. There are expansive options in bioprinting technology that have become more refined and specialized over the years, and stem cells address many limitations in cell source, expansion, and development of bioengineered tissue constructs. While bioprinted stem cells present an opportunity to replicate physiological microenvironments with precision, the future of this practice relies heavily on the optimization of the cellular microenvironment. To fabricate tissue constructs that are useful in replicating physiological conditions in laboratory settings, or in preparation for transplantation to a living host, the microenvironment must mimic conditions that allow bioprinted stem cells to proliferate, differentiate, and migrate. The advances of bioprinting stem cells and directing cell fate have the potential to provide feasible and translatable approach to creating complex tissues and organs. This review will examine the methods through which bioprinted stem cells are differentiated into desired cell lineages through biochemical, biological, and biomechanical techniques.
Collapse
Affiliation(s)
- Lauren N. West-Livingston
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Jihoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
89
|
Fonseca AC, Melchels FPW, Ferreira MJS, Moxon SR, Potjewyd G, Dargaville TR, Kimber SJ, Domingos M. Emulating Human Tissues and Organs: A Bioprinting Perspective Toward Personalized Medicine. Chem Rev 2020; 120:11128-11174. [PMID: 32937071 PMCID: PMC7645917 DOI: 10.1021/acs.chemrev.0c00342] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The lack of in vitro tissue and organ models capable of mimicking human physiology severely hinders the development and clinical translation of therapies and drugs with higher in vivo efficacy. Bioprinting allow us to fill this gap and generate 3D tissue analogues with complex functional and structural organization through the precise spatial positioning of multiple materials and cells. In this review, we report the latest developments in terms of bioprinting technologies for the manufacturing of cellular constructs with particular emphasis on material extrusion, jetting, and vat photopolymerization. We then describe the different base polymers employed in the formulation of bioinks for bioprinting and examine the strategies used to tailor their properties according to both processability and tissue maturation requirements. By relating function to organization in human development, we examine the potential of pluripotent stem cells in the context of bioprinting toward a new generation of tissue models for personalized medicine. We also highlight the most relevant attempts to engineer artificial models for the study of human organogenesis, disease, and drug screening. Finally, we discuss the most pressing challenges, opportunities, and future prospects in the field of bioprinting for tissue engineering (TE) and regenerative medicine (RM).
Collapse
Affiliation(s)
- Ana Clotilde Fonseca
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Ferry P. W. Melchels
- Institute
of Biological Chemistry, Biophysics and Bioengineering, School of
Engineering and Physical Sciences, Heriot-Watt
University, Edinburgh EH14 4AS, U.K.
| | - Miguel J. S. Ferreira
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| | - Samuel R. Moxon
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Geoffrey Potjewyd
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Tim R. Dargaville
- Institute
of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Queensland 4001, Australia
| | - Susan J. Kimber
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Marco Domingos
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| |
Collapse
|
90
|
Costa JB, Park J, Jorgensen AM, Silva-Correia J, Reis RL, Oliveira JM, Atala A, Yoo JJ, Lee SJ. 3D Bioprinted Highly Elastic Hybrid Constructs for Advanced Fibrocartilaginous Tissue Regeneration. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2020; 32:8733-8746. [PMID: 34295019 PMCID: PMC8294671 DOI: 10.1021/acs.chemmater.0c03556] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Advanced strategies to bioengineer a fibrocartilaginous tissue to restore the function of the meniscus are necessary. Currently, 3D bioprinting technologies have been employed to fabricate clinically relevant patient-specific complex constructs to address unmet clinical needs. In this study, a highly elastic hybrid construct for fibrocartilaginous regeneration is produced by co-printing a cell-laden gellan gum/fibrinogen (GG/FB) composite bioink together with a silk fibroin methacrylate (Sil-MA) bioink in an interleaved crosshatch pattern. We characterize each bioink formulation by measuring the rheological properties, swelling ratio, and compressive mechanical behavior. For in vitro biological evaluations, porcine primary meniscus cells (pMCs) are isolated and suspended in the GG/FB bioink for the printing process. The results show that the GG/FB bioink provides a proper cellular microenvironment for maintaining the cell viability and proliferation capacity, as well as the maturation of the pMCs in the bioprinted constructs, while the Sil-MA bioink offers excellent biomechanical behavior and structural integrity. More importantly, this bioprinted hybrid system shows the fibrocartilaginous tissue formation without a dimensional change in a mouse subcutaneous implantation model during the 10-week postimplantation. Especially, the alignment of collagen fibers is achieved in the bioprinted hybrid constructs. The results demonstrate this bioprinted mechanically reinforced hybrid construct offers a versatile and promising alternative for the production of advanced fibrocartilaginous tissue.
Collapse
Affiliation(s)
- João B. Costa
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Jihoon Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Adam M. Jorgensen
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Joana Silva-Correia
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B’s Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
- Corresponding authors. Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States., James J. Yoo, MD, PhD and Sang Jin Lee, PhD, (J. J. Yoo), (S. J. Lee)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
- Corresponding authors. Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States., James J. Yoo, MD, PhD and Sang Jin Lee, PhD, (J. J. Yoo), (S. J. Lee)
| |
Collapse
|
91
|
Tytgat L, Dobos A, Markovic M, Van Damme L, Van Hoorick J, Bray F, Thienpont H, Ottevaere H, Dubruel P, Ovsianikov A, Van Vlierberghe S. High-Resolution 3D Bioprinting of Photo-Cross-linkable Recombinant Collagen to Serve Tissue Engineering Applications. Biomacromolecules 2020; 21:3997-4007. [PMID: 32841006 PMCID: PMC7556543 DOI: 10.1021/acs.biomac.0c00386] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/11/2020] [Indexed: 12/15/2022]
Abstract
Various biopolymers, including gelatin, have already been applied to serve a plethora of tissue engineering purposes. However, substantial concerns have arisen related to the safety and the reproducibility of these materials due to their animal origin and the risk associated with pathogen transmission as well as batch-to-batch variations. Therefore, researchers have been focusing their attention toward recombinant materials that can be produced in a laboratory with full reproducibility and can be designed according to specific needs (e.g., by introducing additional RGD sequences). In the present study, a recombinant protein based on collagen type I (RCPhC1) was functionalized with photo-cross-linkable methacrylamide (RCPhC1-MA), norbornene (RCPhC1-NB), or thiol (RCPhC1-SH) functionalities to enable high-resolution 3D printing via two-photon polymerization (2PP). The results indicated a clear difference in 2PP processing capabilities between the chain-growth-polymerized RCPhC1-MA and the step-growth-polymerized RCPhC1-NB/SH. More specifically, reduced swelling-related deformations resulting in a superior CAD-CAM mimicry were obtained for the RCPhC1-NB/SH hydrogels. In addition, RCPhC1-NB/SH allowed the processing of the material in the presence of adipose tissue-derived stem cells that survived the encapsulation process and also were able to proliferate when embedded in the printed structures. As a consequence, it is the first time that successful HD bioprinting with cell encapsulation is reported for recombinant hydrogel bioinks. Therefore, these results can be a stepping stone toward various tissue engineering applications.
Collapse
Affiliation(s)
- Liesbeth Tytgat
- Brussels
Photonics (B-PHOT) − Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
- Polymer
Chemistry & Biomaterials Group − Centre of Macromolecular
Chemistry (CMaC) − Department of Organic and Macromolecular
Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Agnes Dobos
- 3D Printing
and Biofabrication Group, Institute of Materials
Science and Technology, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
- Austrian
Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Marica Markovic
- 3D Printing
and Biofabrication Group, Institute of Materials
Science and Technology, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
- Austrian
Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Lana Van Damme
- Polymer
Chemistry & Biomaterials Group − Centre of Macromolecular
Chemistry (CMaC) − Department of Organic and Macromolecular
Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Jasper Van Hoorick
- Brussels
Photonics (B-PHOT) − Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
- Polymer
Chemistry & Biomaterials Group − Centre of Macromolecular
Chemistry (CMaC) − Department of Organic and Macromolecular
Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Fabrice Bray
- Miniaturisation
pour l’Analyse, la Synthèse et la Protéomique,
USR 3290 Centre National de la Recherche Scientifique, University of Lille, Villeneuve d’Ascq, 59650 France
| | - Hugo Thienpont
- Brussels
Photonics (B-PHOT) − Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
| | - Heidi Ottevaere
- Brussels
Photonics (B-PHOT) − Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
| | - Peter Dubruel
- Polymer
Chemistry & Biomaterials Group − Centre of Macromolecular
Chemistry (CMaC) − Department of Organic and Macromolecular
Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Aleksandr Ovsianikov
- 3D Printing
and Biofabrication Group, Institute of Materials
Science and Technology, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria
- Austrian
Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Sandra Van Vlierberghe
- Brussels
Photonics (B-PHOT) − Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
- Polymer
Chemistry & Biomaterials Group − Centre of Macromolecular
Chemistry (CMaC) − Department of Organic and Macromolecular
Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| |
Collapse
|
92
|
Koçak E, Yıldız A, Acartürk F. Three dimensional bioprinting technology: Applications in pharmaceutical and biomedical area. Colloids Surf B Biointerfaces 2020; 197:111396. [PMID: 33075661 DOI: 10.1016/j.colsurfb.2020.111396] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022]
Abstract
3D bioprinting is a technology based on the principle of three-dimensional printing of designed biological materials, which has been widely used recently. The production of biological materials, such as tissues, organs, cells and blood vessels with this technology is alternative and promising approach for organ and tissue transplantation. Apart from tissue and organ printing, it has a wide range of usage, such as in vitro/in vivo modeling, production of drug delivery systems and, drug screening. However, there are various restrictions on the use of this technology. In this review, the process steps, classification, advantages, limitations, usage and application areas of 3D bioprinting technology, materials and auxiliary materials used in this technology are discussed.
Collapse
Affiliation(s)
- Esen Koçak
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Gazi University, Ankara, Turkey
| | - Ayşegül Yıldız
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Gazi University, Ankara, Turkey
| | - Füsun Acartürk
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Gazi University, Ankara, Turkey.
| |
Collapse
|
93
|
Belk L, Tellisi N, Macdonald H, Erdem A, Ashammakhi N, Pountos I. Safety Considerations in 3D Bioprinting Using Mesenchymal Stromal Cells. Front Bioeng Biotechnol 2020; 8:924. [PMID: 33154961 PMCID: PMC7588840 DOI: 10.3389/fbioe.2020.00924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 07/17/2020] [Indexed: 12/26/2022] Open
Abstract
Three-dimensional (3D) bioprinting has demonstrated great potential for the fabrication of biomimetic human tissues and complex graft materials. This technology utilizes bioinks composed of cellular elements placed within a biomaterial. Mesenchymal stromal cells (MSCs) are an attractive option for cell selection in 3D bioprinting. MSCs can be isolated from a variety of tissues, can pose vast proliferative capacity and can differentiate to multiple committed cell types. Despite their promising properties, the use of MSCs has been associated with several drawbacks. These concerns are related to the ex vivo manipulation throughout the process of 3D bioprinting. The herein manuscript aims to present the current evidence surrounding these events and propose ways to minimize the risks to the patients following widespread expansion of 3D bioprinting in the medical field.
Collapse
Affiliation(s)
- Lucy Belk
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Nazzar Tellisi
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
- School of Medicine, University of Leeds, Leeds, United Kingdom
- Chapel Allerton Hospital, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Hamish Macdonald
- Gloucester Royal Hospital, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, United Kingdom
| | - Ahmet Erdem
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Chemistry, Kocaeli University, Kocaeli, Turkey
- Department of Biomedical Engineering, Kocaeli University, Kocaeli, Turkey
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
- School of Medicine, University of Leeds, Leeds, United Kingdom
- Chapel Allerton Hospital, Leeds Teaching Hospitals, Leeds, United Kingdom
| |
Collapse
|
94
|
Adhikari J, Roy A, Das A, Ghosh M, Thomas S, Sinha A, Kim J, Saha P. Effects of Processing Parameters of 3D Bioprinting on the Cellular Activity of Bioinks. Macromol Biosci 2020; 21:e2000179. [PMID: 33017096 DOI: 10.1002/mabi.202000179] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022]
Abstract
In this review, few established cell printing techniques along with their parameters that affect the cell viability during bioprinting are considered. 3D bioprinting is developed on the principle of additive manufacturing using biomaterial inks and bioinks. Different bioprinting methods impose few challenges on cell printing such as shear stress, mechanical impact, heat, laser radiation, etc., which eventually lead to cell death. These factors also cause alteration of cells phenotype, recoverable or irrecoverable damages to the cells. Such challenges are not addressed in detail in the literature and scientific reports. Hence, this review presents a detailed discussion of several cellular bioprinting methods and their process-related impacts on cell viability, followed by probable mitigation techniques. Most of the printable bioinks encompass cells within hydrogel as scaffold material to avoid the direct exposure of the harsh printing environment on cells. However, the advantages of printing with scaffold-free cellular aggregates over cell-laden hydrogels have emerged very recently. Henceforth, optimal and favorable crosslinking mechanisms providing structural rigidity to the cell-laden printed constructs with ideal cell differentiation and proliferation, are discussed for improved understanding of cell printing methods for the future of organ printing and transplantation.
Collapse
Affiliation(s)
- Jaideep Adhikari
- J. Adhikari, A. Das, Dr. A. Sinha, M. N. Dastur School of Materials Science and Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Avinava Roy
- A. Roy, Dr. M. Ghosh, Department of Metallurgy and Materials Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Anindya Das
- J. Adhikari, A. Das, Dr. A. Sinha, M. N. Dastur School of Materials Science and Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Manojit Ghosh
- A. Roy, Dr. M. Ghosh, Department of Metallurgy and Materials Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Sabu Thomas
- Prof. S. Thomas, School of Chemical Sciences, MG University, Kottayam, Kerala, 686560, India
| | - Arijit Sinha
- J. Adhikari, A. Das, Dr. A. Sinha, M. N. Dastur School of Materials Science and Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Jinku Kim
- Prof. J. Kim, Department of Bio and Chemical Engineering, Hongik University, Sejong, 30016, South Korea
| | - Prosenjit Saha
- Dr. P. Saha, Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research (JISIASR) Kolkata, JIS University, Arch Water Front Building, Salt Lake City, Kolkata, 700091, India
| |
Collapse
|
95
|
Abdollahiyan P, Oroojalian F, Mokhtarzadeh A, Guardia M. Hydrogel‐Based 3D Bioprinting for Bone and Cartilage Tissue Engineering. Biotechnol J 2020; 15:e2000095. [DOI: 10.1002/biot.202000095] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/22/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Parinaz Abdollahiyan
- Immunology Research Center Tabriz University of Medical Sciences Tabriz 5166614731 Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies School of Medicine North Khorasan University of Medical Sciences Bojnurd 7487794149 Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center Tabriz University of Medical Sciences Tabriz 5166614731 Iran
| | - Miguel Guardia
- Department of Analytical Chemistry University of Valencia Dr. Moliner 50 Burjassot Valencia 46100 Spain
| |
Collapse
|
96
|
Caldwell AS, Aguado BA, Anseth KS. Designing Microgels for Cell Culture and Controlled Assembly of Tissue Microenvironments. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1907670. [PMID: 33841061 PMCID: PMC8026140 DOI: 10.1002/adfm.201907670] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 05/04/2023]
Abstract
Micron-sized hydrogels, termed microgels, are emerging as multifunctional platforms that can recapitulate tissue heterogeneity in engineered cell microenvironments. The microgels can function as either individual cell culture units or can be assembled into larger scaffolds. In this manner, individual microgels can be customized for single or multi-cell co-culture applications, or heterogeneous populations can be used as building blocks to create microporous assembled scaffolds that more closely mimic tissue heterogeneities. The inherent versatility of these materials allows user-defined control of the microenvironments, from the order of singly encapsulated cells to entire three-dimensional cell scaffolds. These hydrogel scaffolds are promising for moving towards personalized medicine approaches and recapitulating the multifaceted microenvironments that exist in vivo.
Collapse
Affiliation(s)
- Alexander S. Caldwell
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| | - Brian A. Aguado
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, USA, 80303
- BioFrontiers Institute, University of Colorado – Boulder, USA, 80303
| |
Collapse
|
97
|
Gu Z, Fu J, Lin H, He Y. Development of 3D bioprinting: From printing methods to biomedical applications. Asian J Pharm Sci 2020; 15:529-557. [PMID: 33193859 PMCID: PMC7610207 DOI: 10.1016/j.ajps.2019.11.003] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/22/2019] [Accepted: 11/17/2019] [Indexed: 12/22/2022] Open
Abstract
Biomanufacturing of tissues/organs in vitro is our big dream, driven by two needs: organ transplantation and accurate tissue models. Over the last decades, 3D bioprinting has been widely applied in the construction of many tissues/organs such as skins, vessels, hearts, etc., which can not only lay a foundation for the grand goal of organ replacement, but also be served as in vitro models committed to pharmacokinetics, drug screening and so on. As organs are so complicated, many bioprinting methods are exploited to figure out the challenges of different applications. So the question is how to choose the suitable bioprinting method? Herein, we systematically review the evolution, process and classification of 3D bioprinting with an emphasis on the fundamental printing principles and commercialized bioprinters. We summarize and classify extrusion-based, droplet-based, and photocuring-based bioprinting methods and give some advices for applications. Among them, coaxial and multi-material bioprinting are highlighted and basic principles of designing bioinks are also discussed.
Collapse
Affiliation(s)
- Zeming Gu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
98
|
Zhu H, Yang H, Ma Y, Lu TJ, Xu F, Genin GM, Lin M. Spatiotemporally Controlled Photoresponsive Hydrogels: Design and Predictive Modeling from Processing through Application. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2000639. [PMID: 32802013 PMCID: PMC7418561 DOI: 10.1002/adfm.202000639] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/16/2020] [Indexed: 05/16/2023]
Abstract
Photoresponsive hydrogels (PRHs) are soft materials whose mechanical and chemical properties can be tuned spatially and temporally with relative ease. Both photo-crosslinkable and photodegradable hydrogels find utility in a range of biomedical applications that require tissue-like properties or programmable responses. Progress in engineering with PRHs is facilitated by the development of theoretical tools that enable optimization of their photochemistry, polymer matrices, nanofillers, and architecture. This review brings together models and design principles that enable key applications of PRHs in tissue engineering, drug delivery, and soft robotics, and highlights ongoing challenges in both modeling and application.
Collapse
Affiliation(s)
- Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Haiqian Yang
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical StructuresNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
- MOE Key Laboratory for Multifunctional Materials and StructuresXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Guy M. Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
- Department of Mechanical Engineering & Materials ScienceWashington University in St. LouisSt. LouisMO63130USA
- NSF Science and Technology Center for Engineering MechanobiologyWashington University in St. LouisSt. LouisMO63130USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| |
Collapse
|
99
|
Ayan B, Wu Y, Karuppagounder V, Kamal F, Ozbolat IT. Aspiration-assisted bioprinting of the osteochondral interface. Sci Rep 2020; 10:13148. [PMID: 32753630 PMCID: PMC7403300 DOI: 10.1038/s41598-020-69960-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/22/2020] [Indexed: 11/09/2022] Open
Abstract
Osteochondral defects contain damage to both the articular cartilage and underlying subchon- dral bone, which remains a significant challenge in orthopedic surgery. Layered structure of bone, cartilage and the bone-cartilage interface must be taken into account in the case of biofabrication of the osteochondral (OC) interface. In this study, a dual layered OC interface was bioprinted using a newly developed aspiration-assisted bioprinting (AAB) technique, which has been the first time that scaffold-free bioprinting was applied to OC interface engineering. Tissue spheroids, made of human adipose-derived stem cells (ADSCs), were differentiated in three dimensions (3D) into chondrogenic and osteogenic spheroids, which were confirmed by immunostaining and histology qualitatively, and biochemistry assays and gene expression, quantitatively. Remarkably, the OC interface was bioprinted by accurate positioning of a layer of osteogenic spheroids onto a sacrificial alginate support followed by another layer of chondrogenic spheroids overlaid by the same support. Spheroids in individual zones fused and the maintenance of phenotypes in both zones confirmed the successful biofabrication of the histomorphologically-relevant OC interface. The biofabrication of OC tissue model without the use of polymeric scaffolds unveils great potential not only in regenerative medicine but also in drug testing and disease modeling for osteoarthritis.
Collapse
Affiliation(s)
- Bugra Ayan
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Yang Wu
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Vengadeshprabhu Karuppagounder
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA, 17033, USA
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Sciences, Department of Orthopedics and Re-Habilitation, Penn State University, Hershey, PA, 17033, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA.
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA.
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA.
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA.
- Department of Neurosurgery, Penn State University, Hershey, PA, 17033, USA.
| |
Collapse
|
100
|
Chen J, Huang D, Wang L, Hou J, Zhang H, Li Y, Zhong S, Wang Y, Wu Y, Huang W. 3D bioprinted multiscale composite scaffolds based on gelatin methacryloyl (GelMA)/chitosan microspheres as a modular bioink for enhancing 3D neurite outgrowth and elongation. J Colloid Interface Sci 2020; 574:162-173. [DOI: 10.1016/j.jcis.2020.04.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
|