1
|
Lan Z, Fletcher A, Bender EC, Huang W, Suggs LJ, Cosgriff-Hernandez E. Hydrogel foam dressings with angiogenic and immunomodulatory factors from mesenchymal stem cells. J Biomed Mater Res A 2024; 112:1388-1398. [PMID: 38270241 DOI: 10.1002/jbm.a.37678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Stem cell therapy and skin substitutes address the stalled healing of chronic wounds in order to promote wound closure; however, the high cost and regulatory hurdles of these treatments limit patient access. A low-cost method to induce bioactive healing has the potential to substantially improve patient care and prevent wound-induced limb loss. A previous study reported that bioactive factors derived from apoptotic-like mesenchymal stem cells (MSCs) demonstrated anti-inflammatory and proangiogenic effects and improved ischemic muscle regeneration. In this work, these MSC-derived bioactive factors were loaded into a hydrogel foam to harness immunomodulatory and angiogenic properties from MSC components to facilitate chronic wound healing without the high cost and translational challenges of cell therapies. After incorporation of bioactive factors, the hydrogel foam retained high absorbency, moisture retention, and target water vapor transmission rate. High loading efficiency was confirmed and release studies indicated that over 90% of loaded factors were released within 24 h. Ethylene oxide sterilization and 4-week storage did not affect the bioactive factor release profile or physical properties of the hydrogel foam dressing. Bioactivity retention of the released factors was also confirmed for as-sterilized, 4°C-stored, and -20°C-stored bioactive hydrogel foams as determined by relevant gene expression levels in treated pro-inflammatory (M1) macrophages. These results support the use of the bioactive dressings as an off-the-shelf product. Overall, this work reports a new method to achieve a first-line wound dressing with the potential to reduce persistent inflammation and promote angiogenesis in chronic wounds.
Collapse
Affiliation(s)
- Ziyang Lan
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, Texas, USA
| | - Alan Fletcher
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, Texas, USA
| | - Elizabeth C Bender
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, Texas, USA
| | - Wenbai Huang
- School of Physical Education, Jinan University, Guangzhou, China
| | - Laura J Suggs
- Department of Biomedical Engineering, the University of Texas at Austin, Austin, Texas, USA
| | | |
Collapse
|
2
|
Raftery RM, Gonzalez Vazquez AG, Walsh DP, Chen G, Laiva AL, Keogh MB, O'Brien FJ. Mobilizing Endogenous Progenitor Cells Using pSDF1α-Activated Scaffolds Accelerates Angiogenesis and Bone Repair in Critical-Sized Bone Defects. Adv Healthc Mater 2024; 13:e2401031. [PMID: 38850118 DOI: 10.1002/adhm.202401031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/05/2024] [Indexed: 06/09/2024]
Abstract
Mobilizing endogenous progenitor cells to repair damaged tissue in situ has the potential to revolutionize the field of regenerative medicine, while the early establishment of a vascular network will ensure survival of newly generated tissue. In this study, a gene-activated scaffold containing a stromal derived factor 1α plasmid (pSDF1α), a pro-angiogenic gene that is also thought to be involved in the recruitment of mesenchymal stromal cells (MSCs) to sites of injury is described. It is shown that over-expression of SDF1α protein enhanced MSC recruitment and induced vessel-like structure formation by endothelial cells in vitro. When implanted subcutaneously, transcriptomic analysis reveals that endogenous MSCs are recruited and significant angiogenesis is stimulated. Just 1-week after implantation into a calvarial critical-sized bone defect, pSDF1α-activated scaffolds are recruited MSCs and rapidly activate angiogenic and osteogenic programs, upregulating Runx2, Dlx5, and Sp7. At the same time-point, pVEGF-activated scaffolds are recruited a variety of cell types, activating endochondral ossification. The early response induced by both scaffolds leads to complete bridging of the critical-sized bone defects within 4-weeks. The versatile cell-free gene-activated scaffold described in this study is capable of harnessing and enhancing the body's own regenerative capacity and has immense potential in a myriad of applications.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- iEd Hub and Department of Anatomy and Neuroscience, College of Medicine and Health, University College Cork, Cork, T12 CY82, Ireland
| | - Arlyng G Gonzalez Vazquez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| | - David P Walsh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- Translational Research in Nanomedical Devices, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Ashang L Laiva
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Michael B Keogh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| |
Collapse
|
3
|
Abdollahi F, Saghatchi M, Paryab A, Malek Khachatourian A, Stephens ED, Toprak MS, Badv M. Angiogenesis in bone tissue engineering via ceramic scaffolds: A review of concepts and recent advancements. BIOMATERIALS ADVANCES 2024; 159:213828. [PMID: 38479240 DOI: 10.1016/j.bioadv.2024.213828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
Due to organ donor shortages, long transplant waitlists, and the complications/limitations associated with auto and allotransplantation, biomaterials and tissue-engineered models are gaining attention as feasible alternatives for replacing and reconstructing damaged organs and tissues. Among various tissue engineering applications, bone tissue engineering has become a promising strategy to replace or repair damaged bone. We aimed to provide an overview of bioactive ceramic scaffolds in bone tissue engineering, focusing on angiogenesis and the effect of different biofunctionalization strategies. Different routes to angiogenesis, including chemical induction through signaling molecules immobilized covalently or non-covalently, in situ secretion of angiogenic growth factors, and the degradation of inorganic scaffolds, are described. Physical induction mechanisms are also discussed, followed by a review of methods for fabricating bioactive ceramic scaffolds via microfabrication methods, such as photolithography and 3D printing. Finally, the strengths and weaknesses of the commonly used methodologies and future directions are discussed.
Collapse
Affiliation(s)
- Farnoosh Abdollahi
- Department of Dentistry, Kashan University of Medical Science, Kashan, Iran
| | - Mahshid Saghatchi
- School of Metallurgy & Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Amirhosein Paryab
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Emma D Stephens
- Department of Biomedical Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Muhammet S Toprak
- Department of Applied Physics, Biomedical and X-ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden
| | - Maryam Badv
- Department of Biomedical Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Libin Cardiovascular Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
4
|
Lee J, Jung S, Hong HK, Jo H, Rhee S, Jeong YL, Ko J, Cho YB, Jeon NL. Vascularized tissue on mesh-assisted platform (VT-MAP): a novel approach for diverse organoid size culture and tailored cancer drug response analysis. LAB ON A CHIP 2024; 24:2208-2223. [PMID: 38533822 DOI: 10.1039/d3lc01055d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
This study presents the vascularized tissue on mesh-assisted platform (VT-MAP), a novel microfluidic in vitro model that uses an open microfluidic principle for cultivating vascularized organoids. Addressing the gap in 3D high-throughput platforms for drug response analysis, the VT-MAP can host tumor clusters of various sizes, allowing for precise, size-dependent drug interaction assessments. Key features include capability for forming versatile co-culture conditions (EC, fibroblasts and colon cancer organoids) that enhance tumor organoid viability and a perfusable vessel network that ensures efficient drug delivery and maintenance of organoid health. The VT-MAP enables the culture and analysis of organoids across a diverse size spectrum, from tens of microns to several millimeters. The VT-MAP addresses the inconsistencies in traditional organoid testing related to organoid size, which significantly impacts drug response and viability. Its ability to handle various organoid sizes leads to results that more accurately reflect patient-derived xenograft (PDX) models and differ markedly from traditional in vitro well plate-based methods. We introduce a novel image analysis algorithm that allows for quantitative analysis of organoid size-dependent drug responses, marking a significant step forward in replicating PDX models. The PDX sample from a positive responder exhibited a significant reduction in cell viability across all organoid sizes when exposed to chemotherapeutic agents (5-FU, oxaliplatin, and irinotecan), as expected for cytotoxic drugs. In sharp contrast, PDX samples of a negative responder showed little to no change in viability in smaller clusters and only a slight reduction in larger clusters. This differential response, accurately replicated in the VT-MAP, underscores its ability to generate data that align with PDX models and in vivo findings. Its capacity to handle various organoid sizes leads to results that more accurately reflect PDX models and differ markedly from traditional in vitro methods. The platform's distinct advantage lies in demonstrating how organoid size can critically influence drug response, revealing insights into cancer biology previously unattainable with conventional techniques.
Collapse
Affiliation(s)
- Jungseub Lee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Sangmin Jung
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Hye Kyoung Hong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyeonsu Jo
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Stephen Rhee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
| | - Ye-Lin Jeong
- Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Jihoon Ko
- Department of Bionano Technology, Gachon University, Seoul, Republic of Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Seoul, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
- Institute of Advanced Machines and Design, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Kwon H, Lee S, Byun H, Huh SJ, Lee E, Kim E, Lee J, Shin H. Engineering pre-vascularized 3D tissue and rapid vascular integration with host blood vessels via co-cultured spheroids-laden hydrogel. Biofabrication 2024; 16:025029. [PMID: 38447223 DOI: 10.1088/1758-5090/ad30c6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/06/2024] [Indexed: 03/08/2024]
Abstract
Recent advances in regenerative medicine and tissue engineering have enabled the biofabrication of three-dimensional (3D) tissue analogues with the potential for use in transplants and disease modeling. However, the practical use of these biomimetic tissues has been hindered by the challenge posed by reconstructing anatomical-scale micro-vasculature tissues. In this study, we suggest that co-cultured spheroids within hydrogels hold promise for regenerating highly vascularized and innervated tissues, bothin vitroandin vivo. Human adipose-derived stem cells (hADSCs) and human umbilical vein cells (HUVECs) were prepared as spheroids, which were encapsulated in gelatin methacryloyl hydrogels to fabricate a 3D pre-vascularized tissue. The vasculogenic responses, extracellular matrix production, and remodeling depending on parameters like co-culture ratio, hydrogel strength, and pre-vascularization time forin vivointegration with native vessels were then delicately characterized. The co-cultured spheroids with 3:1 ratio (hADSCs/HUVECs) within the hydrogel and with a pliable storage modulus showed the greatest vasculogenic potential, and ultimately formedin vitroarteriole-scale vasculature with a longitudinal lumen structure and a complex vascular network after long-term culturing. Importantly, the pre-vascularized tissue also showed anastomotic vascular integration with host blood vessels after transplantation, and successful vascularization that was positive for both CD31 and alpha-smooth muscle actin covering 18.6 ± 3.6μm2of the luminal area. The described co-cultured spheroids-laden hydrogel can therefore serve as effective platform for engineering 3D vascularized complex tissues.
Collapse
Affiliation(s)
- Hyunseok Kwon
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunjin Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
| | - Jinkyu Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
- BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
6
|
Xu F, Dawson C, Hoare T. Multicellular Layered Nanofibrous Poly(Oligo Ethylene Glycol Methacrylate) (POEGMA)-Based Hydrogel Scaffolds via Reactive Cell Electrospinning. Adv Biol (Weinh) 2023; 7:e2300052. [PMID: 37271858 DOI: 10.1002/adbi.202300052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/06/2023] [Indexed: 06/06/2023]
Abstract
While hydrogels are demonstrated to be effective scaffolds for soft tissue engineering, existing fabrication techniques pose limitations in terms of being able to reproduce both the micro/nanofibrous structures of native extracellular matrix as well as the spatial arrangement of different cell types inherent of more complex tissues. Herein, a reactive cell electrospinning strategy is described using hydrazide and aldehyde-functionalized poly(oligoethylene glycol methacrylate) precursor polymers that can create nanofibrous hydrogel scaffolds with controllable local cell gradients using a sequential all-aqueous process that does not require additives or external energy. Cells can be encapsulated directly during the fabrication process in different layers within the scaffold, enabling localized segregation of different cell types within the structures without compromising their capacity to proliferate (≈4-fold increase in cell density over a 14 day incubation period). This sequential reactive electrospinning approach thus offers promise to generate coculture fibrous hydrogel networks in which both the nanoscale architecture and the cell distribution can be controlled, as it is essential to recreate more complex types of tissues.
Collapse
Affiliation(s)
- Fei Xu
- Department of Chemical Engineering, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Chloe Dawson
- Department of Chemical Engineering, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
7
|
Fadl A, Leask A. Hiding in Plain Sight: Human Gingival Fibroblasts as an Essential, Yet Overlooked, Tool in Regenerative Medicine. Cells 2023; 12:2021. [PMID: 37626831 PMCID: PMC10453328 DOI: 10.3390/cells12162021] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Adult human gingival fibroblasts (HGFs), the most abundant cells in the oral cavity, are essential for maintaining oral homeostasis. Compared with other tissues, adult oral mucosal wounds heal regeneratively, without scarring. Relative to fibroblasts from other locations, HGFs are relatively refractory to myofibroblast differentiation, immunomodulatory, highly regenerative, readily obtained via minimally invasive procedures, easily and rapidly expanded in vitro, and highly responsive to growth factors and cytokines. Consequently, HGFs might be a superior, yet perhaps underappreciated, source of adult mesenchymal progenitor cells to use in tissue engineering and regeneration applications, including the treatment of fibrotic auto-immune connective tissue diseases such as scleroderma. Herein, we highlight in vitro and translational studies that have investigated the regenerative and differentiation potential of HGFs, with the objective of outlining current limitations and inspiring future research that could facilitate translating the regenerative potential of HGFs into the clinic.
Collapse
Affiliation(s)
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK S7N 5A2, Canada;
| |
Collapse
|
8
|
Mathis K, Kohon AI, Black S, Meckes B. Light-Controlled Cell-Cell Assembly Using Photocaged Oligonucleotides. ACS MATERIALS AU 2023; 3:386-393. [PMID: 38090125 PMCID: PMC10347689 DOI: 10.1021/acsmaterialsau.3c00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 09/29/2024]
Abstract
The interactions between heterogeneous cell populations play important roles in dictating various cell behaviors. Cell-cell contact mediates communication through the exchange of signaling molecules, electrical coupling, and direct membrane-linked ligand-receptor interactions. In vitro culturing of multiple cell types with control over their specific arrangement is difficult, especially in three-dimensional (3D) systems. While techniques that allow one to control the arrangement of cells and direct contact between different cell types have been developed that expand upon simple co-culture methods, specific control over heterojunctions that form between cells is not easily accomplished with current methods, such as 3D cell-printing. In this article, DNA-mediated cell interactions are combined with cell-compatible photolithographic approaches to control cell assembly. Specifically, cells are coated with oligonucleotides containing DNA nucleobases that are protected with photocleavable moieties; this coating facilitated light-controlled cell assembly when these cells were mixed with cells coated with complementary oligonucleotides. By combining this technology with digital micromirror devices mounted on a microscope, selective activation of specific cell populations for interactions with other cells was achieved. Importantly, this technique is rapid and uses non-UV light sources. Taken together, this technique opens new pathways for on-demand programming of complex cell structures.
Collapse
Affiliation(s)
- Katelyn Mathis
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Afia Ibnat Kohon
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Stephen Black
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Brian Meckes
- Department
of Biomedical Engineering, University of
North Texas, 3940 N Elm Street, Denton, Texas 76207, United States
- BioDiscovery
Institute, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| |
Collapse
|
9
|
Knabe C, Stiller M, Kampschulte M, Wilbig J, Peleska B, Günster J, Gildenhaar R, Berger G, Rack A, Linow U, Heiland M, Rendenbach C, Koerdt S, Steffen C, Houshmand A, Xiang-Tischhauser L, Adel-Khattab D. A tissue engineered 3D printed calcium alkali phosphate bioceramic bone graft enables vascularization and regeneration of critical-size discontinuity bony defects in vivo. Front Bioeng Biotechnol 2023; 11:1221314. [PMID: 37397960 PMCID: PMC10311449 DOI: 10.3389/fbioe.2023.1221314] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction: Recently, efforts towards the development of patient-specific 3D printed scaffolds for bone tissue engineering from bioactive ceramics have continuously intensified. For reconstruction of segmental defects after subtotal mandibulectomy a suitable tissue engineered bioceramic bone graft needs to be endowed with homogenously distributed osteoblasts in order to mimic the advantageous features of vascularized autologous fibula grafts, which represent the standard of care, contain osteogenic cells and are transplanted with the respective blood vessel. Consequently, inducing vascularization early on is pivotal for bone tissue engineering. The current study explored an advanced bone tissue engineering approach combining an advanced 3D printing technique for bioactive resorbable ceramic scaffolds with a perfusion cell culture technique for pre-colonization with mesenchymal stem cells, and with an intrinsic angiogenesis technique for regenerating critical size, segmental discontinuity defects in vivo applying a rat model. To this end, the effect of differing Si-CAOP (silica containing calcium alkali orthophosphate) scaffold microarchitecture arising from 3D powder bed printing (RP) or the Schwarzwalder Somers (SSM) replica fabrication technique on vascularization and bone regeneration was analyzed in vivo. In 80 rats 6-mm segmental discontinuity defects were created in the left femur. Methods: Embryonic mesenchymal stem cells were cultured on RP and SSM scaffolds for 7d under perfusion to create Si-CAOP grafts with terminally differentiated osteoblasts and mineralizing bone matrix. These scaffolds were implanted into the segmental defects in combination with an arteriovenous bundle (AVB). Native scaffolds without cells or AVB served as controls. After 3 and 6 months, femurs were processed for angio-µCT or hard tissue histology, histomorphometric and immunohistochemical analysis of angiogenic and osteogenic marker expression. Results: At 3 and 6 months, defects reconstructed with RP scaffolds, cells and AVB displayed a statistically significant higher bone area fraction, blood vessel volume%, blood vessel surface/volume, blood vessel thickness, density and linear density than defects treated with the other scaffold configurations. Discussion: Taken together, this study demonstrated that the AVB technique is well suited for inducing adequate vascularization of the tissue engineered scaffold graft in segmental defects after 3 and 6 months, and that our tissue engineering approach employing 3D powder bed printed scaffolds facilitated segmental defect repair.
Collapse
Affiliation(s)
- Christine Knabe
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
| | - Michael Stiller
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
- Department of Prosthodontics, Philipps University Marburg, Marburg, Germany
| | - Marian Kampschulte
- Department of Radiology, Justus Liebig University Giessen, Giessen, Germany
| | - Janka Wilbig
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Barbara Peleska
- Department of Prosthodontics, Philipps University Marburg, Marburg, Germany
| | - Jens Günster
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Renate Gildenhaar
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Georg Berger
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Alexander Rack
- Structure of Materials Group, ESRF (European Synchroton Radiation Facility), Grenoble, France
| | - Ulf Linow
- Department of Biomaterials and Multimodal Processing, Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Rendenbach
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Steffen Koerdt
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudius Steffen
- Department of Oral and Maxillofacial Surgery, Charité University Medical Center Berlin (Charité-Universitätsmedizin Berlin), Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alireza Houshmand
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
| | - Li Xiang-Tischhauser
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
| | - Doaa Adel-Khattab
- Department of Experimental Orofacial Medicine, Philipps University Marburg, Marburg, Germany
- Department of Periodontology, Ain Shams University, Cairo, Egypt
| |
Collapse
|
10
|
Tan G, Chen R, Tu X, Guo L, Guo L, Xu J, Zhang C, Zou T, Sun S, Jiang Q. Research on the osteogenesis and biosafety of ECM–Loaded 3D–Printed Gel/SA/58sBG scaffolds. Front Bioeng Biotechnol 2022; 10:973886. [PMID: 36061449 PMCID: PMC9438739 DOI: 10.3389/fbioe.2022.973886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Employing scaffolds containing cell–derived extracellular matrix (ECM) as an alternative strategy for the regeneration of bone defects has shown prominent advantages. Here, gelatin (Gel), sodium alginate (SA) and 58s bioactive glass (58sBG) were incorporated into deionized water to form ink, which was further fabricated into composite scaffolds by the 3D printing technique. Then, rat aortic endothelial cells (RAOECs) or rat bone mesenchymal stem cells (RBMSCs) were seeded on the scaffolds. After decellularization, two kinds of ECM–loaded scaffolds (RAOECs–ECM scaffold and RBMSCs–ECM scaffold) were obtained. The morphological characteristics of the scaffolds were assessed meticulously by scanning electron microscopy (SEM). In addition, the effects of scaffolds on the proliferation, adhesion, and osteogenic and angiogenic differentiation of RBMSCs were evaluated by Calcein AM staining and reverse transcription polymerase chain reaction (RT–PCR). In vivo, full–thickness bone defects with a diameter of 5 mm were made in the mandibles of Sprague–Dawley (SD) rats to assess the bone regeneration ability and biosafety of the scaffolds at 4, 8 and 16 weeks. The osteogenic and angiogenic potential of the scaffolds were investigated by microcomputed tomography (Micro–CT) and histological analysis. The biosafety of the scaffolds was evaluated by blood biochemical indices and histological staining of the liver, kidney and cerebrum. The results showed that the ECM–loaded scaffolds were successfully prepared, exhibiting interconnected pores and a gel–like ECM distributed on their surfaces. Consistently, in vitro experiments demonstrated that the scaffolds displayed favourable cytocompatibility. In vitro osteogenic differentiation studies showed that scaffolds coated with ECM could significantly increase the expression of osteogenic and angiogenic genes. In addition, the results from mandibular defect repair in vivo revealed that the ECM–loaded scaffolds effectively promoted the healing of bone defects when compared to the pure scaffold. Overall, these findings demonstrate that both RAOECs–ECM scaffold and RBMSCs–ECM scaffold can greatly enhance bone formation with good biocompatibility and thus have potential for clinical application in bone regeneration.
Collapse
Affiliation(s)
- Guozhong Tan
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Rongfeng Chen
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Xinran Tu
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Liyang Guo
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lvhua Guo
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Jingyi Xu
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Chengfei Zhang
- Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ting Zou
- Endodontology, Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shuyu Sun
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Shuyu Sun, ; Qianzhou Jiang,
| | - Qianzhou Jiang
- Department of Endodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
- *Correspondence: Shuyu Sun, ; Qianzhou Jiang,
| |
Collapse
|
11
|
Shahabipour F, Tavafoghi M, Aninwene GE, Bonakdar S, Oskuee RK, Shokrgozar MA, Potyondy T, Alambeigi F, Ahadian S. Coaxial 3D bioprinting of tri-polymer scaffolds to improve the osteogenic and vasculogenic potential of cells in co-culture models. J Biomed Mater Res A 2022; 110:1077-1089. [PMID: 35025130 DOI: 10.1002/jbm.a.37354] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022]
Abstract
The crosstalk between osteoblasts and endothelial cells is critical for bone vascularization and regeneration. Here, we used a coaxial 3D bioprinting method to directly print an osteon-like structure by depositing angiogenic and osteogenic bioinks from the core and shell regions of the coaxial nozzle, respectively. The bioinks were made up of gelatin, gelatin methacryloyl (GelMA), alginate, and hydroxyapatite (HAp) nanoparticles and were loaded with human umbilical vascular endothelial cells (HUVECs) and osteoblasts (MC3T3) in the core and shell regions, respectively. Conventional monoaxial 3D bioprinting was used as a control method, where the hydrogels, HAp nanoparticles, MC3T3 cells, and HUVECs were all mixed in one bioink and printed from the core nozzle. As a result, the bioprinted scaffolds were composed of cell-laden fibers with either a core-shell or homogenous structure, providing a non-contact (indirect) or contact (direct) co-culture of MC3T3 cells and HUVECs, respectively. Both structures supported the 3D culture of HUVECs and osteoblasts over a long period. The scaffolds also supported the expression of osteogenic and angiogenic factors. However, the gene expression was significantly higher for the core-shell structure than the homogeneous structure due to the well-defined distribution of osteoblasts and endothelial cells and the formation of vessel-like structures in the co-culture system. Our results indicated that the coaxial bioprinting technique, with the ability to create a non-contact co-culture of cells, can provide a more efficient bioprinting strategy for printing highly vascularized and bioactive bone structures.
Collapse
Affiliation(s)
- Fahimeh Shahabipour
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Maryam Tavafoghi
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - George E Aninwene
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA.,California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Kazemi Oskuee
- Biomedical Applied Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Tyler Potyondy
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Farshid Alambeigi
- Walker Department of Mechanical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, USA
| |
Collapse
|
12
|
Fang Y, Sun W, Zhang T, Xiong Z. Recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps: A review. Biomaterials 2021; 280:121298. [PMID: 34864451 DOI: 10.1016/j.biomaterials.2021.121298] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The field of cardiac tissue engineering has advanced over the past decades; however, most research progress has been limited to engineered cardiac tissues (ECTs) at the microscale with minimal geometrical complexities such as 3D strips and patches. Although microscale ECTs are advantageous for drug screening applications because of their high-throughput and standardization characteristics, they have limited translational applications in heart repair and the in vitro modeling of cardiac function and diseases. Recently, researchers have made various attempts to construct engineered cardiac pumps (ECPs) such as chambered ventricles, recapitulating the geometrical complexity of the native heart. The transition from microscale ECTs to ECPs at a translatable scale would greatly accelerate their translational applications; however, researchers are confronted with several major hurdles, including geometrical reconstruction, vascularization, and functional maturation. Therefore, the objective of this paper is to review the recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps. We first review the bioengineering approaches to fabricate ECPs, and then emphasize the unmatched potential of 3D bioprinting techniques. We highlight key advances in bioprinting strategies with high cell density as researchers have begun to realize the critical role that the cell density of non-proliferative cardiomyocytes plays in the cell-cell interaction and functional contracting performance. We summarize the current approaches to engineering vasculatures both at micro- and meso-scales, crucial for the survival of thick cardiac tissues and ECPs. We showcase a variety of strategies developed to enable the functional maturation of cardiac tissues, mimicking the in vivo environment during cardiac development. By highlighting state-of-the-art research, this review offers personal perspectives on future opportunities and trends that may bring us closer to the promise of functional ECPs.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| |
Collapse
|
13
|
Dalisson B, Charbonnier B, Aoude A, Gilardino M, Harvey E, Makhoul N, Barralet J. Skeletal regeneration for segmental bone loss: Vascularised grafts, analogues and surrogates. Acta Biomater 2021; 136:37-55. [PMID: 34626818 DOI: 10.1016/j.actbio.2021.09.053] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023]
Abstract
Massive segmental bone defects (SBD) are mostly treated by removing the fibula and transplanting it complete with blood supply. While revolutionary 50 years ago, this remains the standard treatment. This review considers different strategies to repair SBD and emerging potential replacements for this highly invasive procedure. Prior to the technical breakthrough of microsurgery, researchers in the 1960s and 1970s had begun to make considerable progress in developing non autologous routes to repairing SBD. While the breaktthrough of vascularised bone transplantation solved the immediate problem of a lack of reliable repair strategies, much of their prior work is still relevant today. We challenge the assumption that mimicry is necessary or likely to be successful and instead point to the utility of quite crude (from a materials technology perspective), approaches. Together there are quite compelling indications that the body can regenerate entire bone segments with few or no exogenous factors. This is important, as there is a limit to how expensive a bone repair can be and still be widely available to all patients since cost restraints within healthcare systems are not likely to diminish in the near future. STATEMENT OF SIGNIFICANCE: This review is significant because it is a multidisciplinary view of several surgeons and scientists as to what is driving improvement in segmental bone defect repair, why many approaches to date have not succeeded and why some quite basic approaches can be as effective as they are. While there are many reviews of the literature of grafting and bone repair the relative lack of substantial improvement and slow rate of progress in clinical translation is often overlooked and we seek to challenge the reader to consider the issue more broadly.
Collapse
|
14
|
Bone morphogenetic protein 9 enhances osteogenic and angiogenic responses of human amniotic mesenchymal stem cells cocultured with umbilical vein endothelial cells through the PI3K/AKT/m-TOR signaling pathway. Aging (Albany NY) 2021; 13:24829-24849. [PMID: 34837694 PMCID: PMC8660623 DOI: 10.18632/aging.203718] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/25/2021] [Indexed: 01/06/2023]
Abstract
Background: Neovascularization plays an essential part in bone fracture and defect healing, constructing tissue engineered bone that targets bone regeneration. Bone morphogenetic protein 9 (BMP9) is a regular indicator that potentiates osteogenic and angiogenic differentiation of MSCs. Objectives: To investigate the effects of BMP9 on osteogenesis and angiogenesis of human amniotic mesenchymal stem cells (hAMSCs) cocultured with human umbilical vein endothelial cells (HUVECs) and determine the possible underlying molecular mechanism. Results: The isolated hAMSCs expressed surface markers of MSCs. hAMSCs cocultured with HUVECs enhance osteogenic differentiation and upregulate the expression of angiogenic factors. BMP9 not only potentiates angiogenic signaling of hAMSCs cocultured with HUVECs also increases ectopic bone formation and subcutaneous vessel invasion. Mechanically, the coupling effect between osteogenesis and angiogenesis induced by BMP9 was activated by the BMP/Smad and PI3K/AKT/m-TOR signaling pathways. Conclusions: BMP9-enhanced osteoblastic and angiogenic differentiation in cocultivation with hAMSCs and HUVECs in vitro and in vivo also provide a chance to harness the BMP9-regulated coordinated effect between osteogenic and angiogenic pathways through BMP/Smad and PI3K/AKT/m-TOR signalings. Materials and Methods: The ALP and Alizarin Red S staining assay to determine the effects of osteoblastic differentiation. RT-qPCR and western blot was measured the expression of angiogenesis-related factors. Ectopic bone formation was established and retrieved bony masses were subjected to histochemical staining. The angiogenesis ability and vessel invasion were subsequently determined by immunofluorescence staining. Molecular mechanisms such as the BMP/Smad and PI3K/AKT/m-TOR signaling pathways were detected by ELISA and western blot analysis.
Collapse
|
15
|
Sheng L, Li M, Zheng S, Qi J. Adjusting the accuracy of PEGDA-GelMA vascular network by dark pigments via digital light processing printing. J Biomater Appl 2021; 36:1173-1187. [PMID: 34738507 DOI: 10.1177/08853282211053081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascularization is one of the most important factors greatly influencing scaffold regeneration. In this study, a precise network of hollow vessels was printed by digital light processing (DLP) with poly(ethylene glycol) diacrylate (PEGDA)/gelatin-methacryloyl (GelMA), and dark pigmentation absorbers were added to ensure printing accuracy. First, the compound bio-inks of the PEGDA-GelMA hydrogel were prepared for direct vascular printing, and a high-precision DLP system was established. Second, the printing effects of three dark absorbers, namely, nigrosin, brilliant black, and brilliant blue, on the x-, y-, and z-axes were studied. By printing models with different densities, it was determined that 0.2% nigrosin, 0.1% brilliant black, and 0.3% brilliant blue had better effects on the x- and y-axes accuracy, and the absorbance of the absorbers played a decisive role in adjusting the accuracy. Additionally, to solve the problem of uneven curing on the upper and lower surfaces caused by the addition of an absorber with high absorbance, a model of the difference in curing width between the upper and lower surfaces of a unit-layer slice based on high-absorbance absorbers was established, and the reference value for the slice thickness was calculated. Third, the biological and mechanical properties of the bio-inks were verified with scanning electron microscopy and Fourier transform infrared, and by tensile, swelling, degradation, and cytotoxicity tests on different concentrations of PEGDA-GelMA hydrogel and absorbers. The results showed that 30% PEGDA-7% GelMA/0.1% brilliant black was the optimal preparation to print a hollow vascular network. The error of the printing tube wall and cavity was between 1% and 3%, which demonstrates the high precision of the method. Human umbilical vein endothelial cells were planted in the lumen, and the survival rate achieved 107% on the seventh day, demonstrating the good biocompatibility of the composite hydrogel.
Collapse
Affiliation(s)
- Lin Sheng
- 12605Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin, China
| | - Mo Li
- 12605Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin, China
| | - Shuxian Zheng
- 12605Tianjin Key Laboratory of Equipment Design and Manufacturing Technology, School of Mechanical Engineering, Tianjin University, Tianjin, China
| | - Jian Qi
- 66270School of Mechanical Engineering, Tianjin University of Technology and Education, China
| |
Collapse
|
16
|
Tabatabaei F, Rasoulianboroujeni M, Yadegari A, Tajik S, Moharamzadeh K, Tayebi L. Osteo-mucosal engineered construct: In situ adhesion of hard-soft tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112255. [PMID: 34474817 DOI: 10.1016/j.msec.2021.112255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The aim of this work was to combine engineered hard and soft tissue, adopting a new method for interfacial adhesion of osteo-mucosal construct. We hypothesized that the chemical procedure involved in this method not only adheres the components, but also improves the cell growth inside them. METHODS 3D-printed functionally-graded porous hard-tissue scaffolds were characterized, functionalized by aminolysis and tyrosinase, and accommodated by human osteoblast cells. Introducing amino groups through aminolysis and inducing dopaquinones by tyrosinase can take part in the Michael additions to cause the adhesion. Subsequently, fully-differentiated engineered oral mucosa was formed directly on the surface of hard tissue. Constructs were assessed in term of morphology, structure, chemical composition, histology, and cytocompatibility. Interfacial adhesion was compared to a control group prepared by using a biological glue for the attachment of the soft and hard tissues. RESULTS The data confirmed higher proliferation of osteoblast cells via aminolysis and improved osteoblast cells distribution and differentiation by incorporation of tyrosinase in collagen. There was evidence of multilayered, stratified epithelium on the osteo-mucosal model with viable fibroblasts and osteoblasts within the lamina propria and bone tissue layers. Our method of adhesion resulted in cohesive debonding within the engineered soft tissue; while in the control group, adhesive debonding and complete separation of the oral mucosa from the hard tissue was observed. Although the shear strength of the osteo-mucosal model (157.6 kDa ± 25.1) was slightly higher than that of the control group (149.4 kDa ± 23.1), there was no statistically significant difference between them (p > 0.05). However, the advantage of our in situ adhesion approach is the absence of a barrier like glue which can disrupt direct cellular communications between tissues. SIGNIFICANCE This study provides a novel method of directly combining tissue-engineered human bone with oral mucosa, which has the potential to improve cell-ingrowth and tissue integration. This engineered tissue construct, after further optimization, can be used clinically as a graft material in various oral surgeries and can also be employed as an in vitro model to investigate many aspects of oral diseases and examine dental materials and oral health care products as a replacement of in vivo models.
Collapse
Affiliation(s)
| | | | - Amir Yadegari
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Sanaz Tajik
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine (HBMCDM), Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, United Arab Emirates; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA.
| |
Collapse
|
17
|
Ma J, Qin C, Wu J, Zhang H, Zhuang H, Zhang M, Zhang Z, Ma L, Wang X, Ma B, Chang J, Wu C. 3D Printing of Strontium Silicate Microcylinder-Containing Multicellular Biomaterial Inks for Vascularized Skin Regeneration. Adv Healthc Mater 2021; 10:e2100523. [PMID: 33963672 DOI: 10.1002/adhm.202100523] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/27/2021] [Indexed: 12/12/2022]
Abstract
The reconstruction of dermal blood vessels is essential for skin regeneration process. However, the lack of vascular structure, insufficient angiogenesis induction, and ineffective graft-host anastomosis of the existing skin substitutes are major bottle-necks for permanent skin replacement in tissue engineering. In this study, the uniform strontium silicate (SS) microcylinders are successfully synthesized and integrated into the biomaterial ink to serve as stable cell-induced factors for angiogenesis, and then a functional skin substitute based on a vascularization-induced biomimetic multicellular system is prepared via a "cell-writing" bioprinting technology. With an unprecedented combination of vascularized skin-mimicking structure and vascularization-induced function, the SS-containing multicellular system exhibits outstanding angiogenic activity both in vitro and in vivo. As a result, the bioprinted skin substitutes significantly accelerate the healing of both acute and chronic wounds by promoting the graft-host integration and vascularized skin regeneration in three animal models. Therefore, the study provides a referable strategy to fabricate biomimetic multicellular constructs with angiogenesis-induced function for regeneration of vascularized complex and hierarchical tissues.
Collapse
Affiliation(s)
- Jingge Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Chen Qin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jinfu Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Hongjian Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Hui Zhuang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Meng Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Zhaowenbin Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Lingling Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Xin Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Bing Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| |
Collapse
|
18
|
Masson-Meyers DS, Tayebi L. Vascularization strategies in tissue engineering approaches for soft tissue repair. J Tissue Eng Regen Med 2021; 15:747-762. [PMID: 34058083 DOI: 10.1002/term.3225] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Insufficient vascularization during tissue repair is often associated with poor clinical outcomes. This is a concern especially when patients have critical-sized injuries, where the size of the defect restricts vascularity, or even in small defects that have to be treated under special conditions, such as after radiation therapy (relevant to tumor resection) that hinders vascularity. In fact, poor vascularization is one of the major obstacles for clinical application of tissue engineering methods in soft tissue repair. As a key issue, lack of graft integration, caused by inadequate vascularization after implantation, can lead to graft failure. Moreover, poor vascularization compromises the viability of cells seeded in deep portions of scaffolds/graft materials, due to hypoxia and insufficient nutrient supply. In this article we aim to review vascularization strategies employed in tissue engineering techniques to repair soft tissues. For this purpose, we start by providing a brief overview of the main events during the physiological wound healing process in soft tissues. Then, we discuss how tissue repair can be achieved through tissue engineering, and considerations with regards to the choice of scaffold materials, culture conditions, and vascularization techniques. Next, we highlight the importance of vascularization, along with strategies and methods of prevascularization of soft tissue equivalents, particularly cell-based prevascularization. Lastly, we present a summary of commonly used in vitro methods during the vascularization of tissue-engineered soft tissue constructs.
Collapse
Affiliation(s)
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| |
Collapse
|
19
|
Jablonská E, Horkavcová D, Rohanová D, Brauer DS. A review of in vitro cell culture testing methods for bioactive glasses and other biomaterials for hard tissue regeneration. J Mater Chem B 2021; 8:10941-10953. [PMID: 33169773 DOI: 10.1039/d0tb01493a] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bioactive glasses are used to regenerate bone by a mechanism which involves surface degradation, the release of ions such as calcium, soluble silica and phosphate and the precipitation of a biomimetic apatite surface layer on the glass. One major area of bioactive glass research is the incorporation of therapeutically active ions to broaden the application range of these materials. When developing such new compositions, in vitro cell culture studies are a key part of their characterisation. However, parameters of cell culture studies vary widely, and depending on the intended use of bioactive glass compositions, different layouts, cell types and assays need to be used. The aim of this publication is to provide materials scientists, particularly those new to cell culture studies, with a tool for selecting the most appropriate assays to give insight into the properties of interest.
Collapse
Affiliation(s)
- Eva Jablonská
- Laboratory of Molecular Biology and Virology, Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technická 3, 166 28 Prague 6, Czech Republic.
| | - Diana Horkavcová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Dana Rohanová
- Laboratory of Chemistry and Technology of Glasses, Department of Glass and Ceramics, University of Chemistry and Technology, Technická 5, 166 28 Prague 6, Czech Republic
| | - Delia S Brauer
- Otto Schott Institute of Materials Research, Faculty of Chemistry and Earth Sciences, Friedrich Schiller University Jena, Fraunhoferstr. 6, 07743 Jena, Germany.
| |
Collapse
|
20
|
Dobos A, Gantner F, Markovic M, Van Hoorick J, Tytgat L, Van Vlierberghe S, Ovsianikov A. On-chip high-definition bioprinting of microvascular structures. Biofabrication 2021; 13:015016. [PMID: 33586666 DOI: 10.1088/1758-5090/abb063] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
'Organ-on-chip' devices which integrate three-dimensional (3D) cell culture techniques with microfluidic approaches have the capacity to overcome the limitations of classical 2D platforms. Although several different strategies have been developed to improve the angiogenesis within hydrogels, one of the main challenges in tissue engineering remains the lack of vascularization in the fabricated 3D models. The present work focuses on the high-definition (HD) bioprinting of microvascular structures directly on-chip using two-photon polymerization (2PP). 2PP is a nonlinear process, where the near-infrared laser irradiation will only lead to the polymerization of a very small volume pixel (voxel), allowing the fabrication of channels in the microvascular range (10-30 µm in diameter). Additionally, 2PP not only enables the fabrication of sub-micrometer resolution scaffolds but also allows the direct embedding of cells within the produced structure. The accuracy of the 2PP printing parameters were optimized in order to achieve high-throughput and HD production of microfluidic vessel-on-chip platforms. The spherical aberrations stemming from the refractive index mismatch and the focusing depth inside the sample were simulated and the effect of the voxel compensation as well as different printing modes were demonstrated. Different layer spacings and their dependency on the applied laser power were compared both in terms of accuracy and required printing time resulting in a 10-fold decrease in structuring time while yielding well-defined channels of small diameters. Finally, the capacity of 2PP to create vascular structures within a microfluidic chip was tested with two different settings, by direct embedding of a co-culture of endothelial- and supporting cells during the printing process and by creating a supporting, cell-containing vascular scaffold barrier where the endothelial cell spheroids can be seeded afterwards. The functionality of the formed vessels was demonstrated with immunostaining of vascular endothelial cadherin (VE-Cadherin) endothelial adhesion molecules in both static and perfused culture.
Collapse
Affiliation(s)
- Agnes Dobos
- 3D Printing and Biofabrication Group, Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria. Austrian Cluster for Tissue Regeneration (http://tissue-regeneration.at), Austria
| | | | | | | | | | | | | |
Collapse
|
21
|
Gonçalves RC, Banfi A, Oliveira MB, Mano JF. Strategies for re-vascularization and promotion of angiogenesis in trauma and disease. Biomaterials 2020; 269:120628. [PMID: 33412374 DOI: 10.1016/j.biomaterials.2020.120628] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022]
Abstract
The maintenance of a healthy vascular system is essential to ensure the proper function of all organs of the human body. While macrovessels have the main role of blood transportation from the heart to all tissues, microvessels, in particular capillaries, are responsible for maintaining tissues' functionality by providing oxygen, nutrients and waste exchanges. Occlusion of blood vessels due to atherosclerotic plaque accumulation remains the leading cause of mortality across the world. Autologous vein and artery grafts bypassing are the current gold standard surgical procedures to substitute primarily obstructed vascular structures. Ischemic scenarios that condition blood supply in downstream tissues may arise from blockage phenomena, as well as from other disease or events leading to trauma. The (i) great demand for new vascular substitutes, arising from both the limited availability of healthy autologous vessels, as well as the shortcomings associated with small-diameter synthetic vascular grafts, and (ii) the challenging induction of the formation of adequate and stable microvasculature are current driving forces for the growing interest in the development of bioinspired strategies to ensure the proper function of vasculature in all its dimensional scales. Here, a critical review of well-established technologies and recent biotechnological advances to substitute or regenerate the vascular system is provided.
Collapse
Affiliation(s)
- Raquel C Gonçalves
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Andrea Banfi
- Department of Biomedicine, University of Basel, Basel, 4056, Switzerland; Department of Surgery, University Hospital Basel, Basel, 4056, Switzerland
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
22
|
Use of in vitro bone models to screen for altered bone metabolism, osteopathies, and fracture healing: challenges of complex models. Arch Toxicol 2020; 94:3937-3958. [PMID: 32910238 PMCID: PMC7655582 DOI: 10.1007/s00204-020-02906-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Approx. every third hospitalized patient in Europe suffers from musculoskeletal injuries or diseases. Up to 20% of these patients need costly surgical revisions after delayed or impaired fracture healing. Reasons for this are the severity of the trauma, individual factors, e.g, the patients’ age, individual lifestyle, chronic diseases, medication, and, over 70 diseases that negatively affect the bone quality. To investigate the various disease constellations and/or develop new treatment strategies, many in vivo, ex vivo, and in vitro models can be applied. Analyzing these various models more closely, it is obvious that many of them have limits and/or restrictions. Undoubtedly, in vivo models most completely represent the biological situation. Besides possible species-specific differences, ethical concerns may question the use of in vivo models especially for large screening approaches. Challenging whether ex vivo or in vitro bone models can be used as an adequate replacement for such screenings, we here summarize the advantages and challenges of frequently used ex vivo and in vitro bone models to study disturbed bone metabolism and fracture healing. Using own examples, we discuss the common challenge of cell-specific normalization of data obtained from more complex in vitro models as one example of the analytical limits which lower the full potential of these complex model systems.
Collapse
|
23
|
Guerreiro SG, Unger RE, Cerqueira NMFSA, Sartoris A, Martins MJ, Barbosa MA, Soares R, Granja PL, Kirkpatrick CJ. Alkaline phosphatase dual-binding sites for collagen dictate cell migration and microvessel assembly in vitro. J Cell Biochem 2020; 122:116-129. [PMID: 32748513 DOI: 10.1002/jcb.29835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 01/01/2023]
Abstract
Interactions between cell types, growth factors, and extracellular matrix components involved in angiogenesis are crucial for new vessel formation leading to tissue regeneration. This study investigated whether cocultures of fibroblasts and endothelial cells (ECs; from macro- or microvasculature) play a role in the formation of microvessel-like structures by ECs, as well as modulate fibroblast differentiation and growth factors production (vascular endothelial cell growth factor, basic fibroblast growth factor, active transforming growth factor-β1, and interleukin-8), which are important for vessel sprouting and maturation. Data obtained revealed that in vitro coculture systems of fibroblasts and human ECs stimulate collagen synthesis and growth factors production by fibroblasts that ultimately affect the formation and distribution of microvessel-like structures in cell cultures. In this study, areas with activated fibroblasts and high alkaline phosphatase (ALP) activity were also observed in cocultures. Molecular docking assays revealed that ALP has two binding positions for collagen, suggesting its impact in collagen proteins' aggregation, cell migration, and microvessel assembly. These findings indicate that bioinformatics and coculture systems are complementary tools for investigating the participation of proteins, like collagen and ALP in angiogenesis.
Collapse
Affiliation(s)
- Susana G Guerreiro
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Ronald E Unger
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Nuno M F S A Cerqueira
- Departamento de Química e Bioquímica, UCIBIO@REQUIMTE, Faculdade de Ciências da Universidade do Porto, Porto, Portugal
| | - Anne Sartoris
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Maria J Martins
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Mário A Barbosa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Raquel Soares
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Pedro L Granja
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Charles J Kirkpatrick
- REPAIR-Lab, Institute of Pathology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
24
|
Smirani R, Rémy M, Devillard R, Naveau A. Engineered Prevascularization for Oral Tissue Grafting: A Systematic Review. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:383-398. [DOI: 10.1089/ten.teb.2020.0093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Rawen Smirani
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, CHU Bordeaux, 33000, Bordeaux, France
| | - Murielle Rémy
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, 33000, Bordeaux, France
| | - Raphael Devillard
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, CHU Bordeaux, 33000, Bordeaux, France
| | - Adrien Naveau
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, CHU Bordeaux, 33000, Bordeaux, France
| |
Collapse
|
25
|
Yu JS, Bagheri N. Agent-Based Models Predict Emergent Behavior of Heterogeneous Cell Populations in Dynamic Microenvironments. Front Bioeng Biotechnol 2020; 8:249. [PMID: 32596213 PMCID: PMC7301008 DOI: 10.3389/fbioe.2020.00249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/10/2020] [Indexed: 01/18/2023] Open
Abstract
Computational models are most impactful when they explain and characterize biological phenomena that are non-intuitive, unexpected, or difficult to study experimentally. Countless equation-based models have been built for these purposes, but we have yet to realize the extent to which rules-based models offer an intuitive framework that encourages computational and experimental collaboration. We develop ARCADE, a multi-scale agent-based model to interrogate emergent behavior of heterogeneous cell agents within dynamic microenvironments and demonstrate how complexity of intracellular metabolism and signaling modules impacts emergent dynamics. We perform in silico case studies on context, competition, and heterogeneity to demonstrate the utility of our model for gaining computational and experimental insight. Notably, there exist (i) differences in emergent behavior between colony and tissue contexts, (ii) linear, non-linear, and multimodal consequences of parameter variation on competition in simulated co-cultures, and (iii) variable impact of cell and population heterogeneity on emergent outcomes. Our extensible framework is easily modified to explore numerous biological systems, from tumor microenvironments to microbiomes.
Collapse
Affiliation(s)
- Jessica S Yu
- Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Neda Bagheri
- Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States.,Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL, United States.,Center for Synthetic Biology, Northwestern University, Evanston, IL, United States.,Biology, University of Washington, Seattle, WA, United States.,Chemical Engineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
26
|
Han G, Zheng Z, Pan Z, Lin Y, Gan S, Jiao Y, Li H, Zhou C, Ding S, Li L. Sulfated chitosan coated polylactide membrane enhanced osteogenic and vascularization differentiation in MC3T3-E1s and HUVECs co-cultures system. Carbohydr Polym 2020; 245:116522. [PMID: 32718626 DOI: 10.1016/j.carbpol.2020.116522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/22/2022]
Abstract
This study aimed to compare the effects of the two type chitosan derivatives, sulfated chitosan (SCS) and phosphorylated chitosan (PCS), coated on poly(d,l-lactide) (PDLLA) membrane via polydopamine, respectively, on vascularization and osteogenesis in vitro. Mouse preosteoblast cells (MC3T3-E1s) and human umbilical vein endothelial cells (HUVECs) were used as co-cultures system. The effects of two type membranes on calcium deposition, alkaline phosphatase (ALP) activity, vascularization related factors nitric oxide (NO) and angiogenic growth factor vascular endothelial growth factor (VEGF) were assessed. The changes of osteogenic and angiogenic related gene, and protein expression were evaluated too. In fact, SCS modified PDLLA membrane had the highest related gene and protein expression than other PDLLA membranes. Our results demonstrated that the SCS maybe a promising matrix for bone regeneration by co-cultures of ECs and OCs than PCS.
Collapse
Affiliation(s)
- Guijuan Han
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Zexiang Zheng
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Zhicheng Pan
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Yucheng Lin
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Shuchun Gan
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China
| | - Yanpeng Jiao
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China
| | - Hong Li
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China
| | - Changren Zhou
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China
| | - Shan Ding
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China.
| | - Lihua Li
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, PR China; Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
27
|
Böhrnsen F, Melsheimer P, Natorp M, Rolf H, Schminke B, Kauffmann P, Wolfer S, Schliephake H. Cotransplantation of mesenchymal stromal cells and endothelial cells on calcium carbonate and hydroxylapatite scaffolds in vivo. J Craniomaxillofac Surg 2020; 49:238-245. [PMID: 33483245 DOI: 10.1016/j.jcms.2020.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/06/2020] [Accepted: 03/08/2020] [Indexed: 10/24/2022] Open
Abstract
This study investigated the cotransplantation of bone marrow mesenchymal stromal cells (BMSC) and human umbilical cord endothelial cells (HUVEC), and evaluated their contribution to vascular and bone tissue engineering in vivo. To evaluate the success of osteogenic differentiation and timely vascularization of different osteoconductive scaffolds in vivo, we transferred BMSC and HUVEC pre-cultivated calcium carbonate (CaCO3) and hydroxylapatite (HA) matrices into immunocompromised RNU-rats, and analyzed mineralization, expression of osteopontin, and vascular integration via new vessel formation. After in vivo transplantation, pre-cultivated scaffolds demonstrated overall improved mineralization of 44% for CaCO3 (p = 0.01, SD ± 14.3) and 34% for HA (p = 0.001, SD ± 17.8), as well as improved vascularization of 5.6 vessels/0.1 mm2 on CaCO3 (p < 0.0001, SD ± 2.0) and 5.3 vessels/0.1 mm2 on HA (p < 0.0001, SD ± 2.4) compared with non-pre-cultivated controls. However, no significant differences between the implantation of BMSC-only, HUVEC-only, or BMSC + HUVEC cocultures could be observed. There is an increasing demand for improved bone regeneration in tissue engineering. Cotransplantation of mesenchymal stromal cells and endothelial cells often demonstrates synergistic improvements in vitro. However, the benefits or superiority of cotransplantation was not evident in vivo and so will require further investigation.
Collapse
Affiliation(s)
- Florian Böhrnsen
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany.
| | - Petra Melsheimer
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Mareike Natorp
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Hans Rolf
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Philipp Kauffmann
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Susanne Wolfer
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| | - Henning Schliephake
- Department of Oral and Maxillofacial Surgery, University Medicine Göttingen, Germany
| |
Collapse
|
28
|
Nilforoushzadeh MA, Sisakht MM, Amirkhani MA, Seifalian AM, Banafshe HR, Verdi J, Nouradini M. Engineered skin graft with stromal vascular fraction cells encapsulated in fibrin–collagen hydrogel: A clinical study for diabetic wound healing. J Tissue Eng Regen Med 2020; 14:424-440. [DOI: 10.1002/term.3003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/18/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022]
Affiliation(s)
| | - Mahsa Mollapour Sisakht
- Skin and Stem Cell Research CenterTehran University of Medical Sciences Tehran Iran
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
| | - Mohammad Amir Amirkhani
- Stem Cell and Regenerative Medicine Center of ExcellenceTehran University of Medical Sciences Tehran Iran
| | - Alexander M. Seifalian
- Nanotechnology and Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd)The London BioScience Innovation Centre London UK
| | - Hamid Reza Banafshe
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
- Physiology Research CenterKashan University of Medical Sciences Kashan Iran
| | - Javad Verdi
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Tehran Iran
| | - Mehdi Nouradini
- Applied Cell Sciences DepartmentKashan University of Medical Science Kashan Iran
| |
Collapse
|
29
|
Zhang X, Simmons CA, Paul Santerre J. Paracrine signalling from monocytes enables desirable extracellular matrix accumulation and temporally appropriate phenotype of vascular smooth muscle cell-like cells derived from adipose stromal cells. Acta Biomater 2020; 103:129-141. [PMID: 31821896 DOI: 10.1016/j.actbio.2019.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022]
Abstract
In vascular tissue engineering, the ability to obtain a robust and safe vascular tissue cell source (e.g. vascular smooth muscle cells (VSMCs)) and to promote vascular tissue-specific extracellular matrix (ECM) protein production is critically important. Mature blood vessel-derived VSMCs are not practical for in vitro vascular tissue regeneration. The authors have conceived a strategy to differentiate adipose derived stromal cells (ASCs) into VSMC-like cells (ASC-VSMCs) that were similar to mature umbilical artery VSMCs at the transcriptional, protein and contraction function levels. Monocytes/macrophages are known as important regulators of the inflammation and regeneration processes within different tissue types of the body. However, our understanding of the potential interactions between specific tissue-like cells differentiated from stem/stromal cells (e.g. ASC-VSMCs) and monocytes/macrophages (cued by specific biomaterial scaffolds) is still limited. In this study, indirect and direct ASC-VSMC-monocyte co-cultures were constructed within a porous polyurethane scaffold (D-PHI) previously shown to have an immunomodulatory character. The effects of monocytes/macrophages on the cellularity (cell number detected with DNA quantification assay), ECM (glycosaminoglycan (GAG), collagen, and elastin) accumulation as well as the maintenance of contractile VSMC markers (calponin and smoothelin) of the ASC-VSMCs after a month of co-culture were investigated. It was found that monocyte paracrine signalling in D-PHI positively affected the cellularity and ECM accumulation of ASC-VSMCs in co-culture. Cause-effect relationships were also identified between the release of pro-inflammatory/anti-inflammatory factors (i.e. IL6, TGF-β1) in co-culture and the expression of contractile proteins (calponin and smoothelin) by ASC-VSMCs. This study demonstrated the importance of combining an immune cell strategy with stromal cell derived VSMCs (i.e. ASC-VSMCs) to achieve a practical vascular tissue engineering outcome. STATEMENT OF SIGNIFICANCE: Adipose stromal cell derived-vascular smooth muscle cells (ASC-VSMCs) are a promising cell source for vascular tissue engineering. Monocytes/monocyte derived macrophages can be harnessed as an immune-assisted strategy to promote vascular tissue regeneration. This study demonstrated that the co-culture of human ASC-VSMCs with monocytes significantly enhanced the cellularity and extracellular matrix (ECM) accumulation within anionic polyurethane (D-PHI) scaffolds, partially mediated by monocyte paracrine signalling mechanisms. In addition, specific VSMC contractile markers (calponin and smoothelin) were still present in ASC-VSMCs when the cells were exposed to monocytes for a month in vitro. This study corroborated the potential selection of ASC-VSMCs for in vitro engineering of vascular tissue in an immunomodulatory biomaterial scaffold (e.g. D-PHI) based co-culture system containing monocytes.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th floor, room 1435, Toronto, Ontario M5G 1M1, Canada
| | - Craig A Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario M5S 3G8, Canada; Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th floor, room 1435, Toronto, Ontario M5G 1M1, Canada
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, 14th floor, room 1435, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
30
|
Quade M, Münch P, Lode A, Duin S, Vater C, Gabrielyan A, Rösen‐Wolff A, Gelinsky M. The Secretome of Hypoxia Conditioned hMSC Loaded in a Central Depot Induces Chemotaxis and Angiogenesis in a Biomimetic Mineralized Collagen Bone Replacement Material. Adv Healthc Mater 2020; 9:e1901426. [PMID: 31830380 DOI: 10.1002/adhm.201901426] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/27/2019] [Indexed: 12/18/2022]
Abstract
The development of biomaterials with intrinsic potential to stimulate endogenous tissue regeneration at the site of injury is a main demand on future implants in regenerative medicine. For critical-sized bone defects, an in situ tissue engineering concept is devised based on biomimetic mineralized collagen scaffolds. These scaffolds are functionalized with a central depot loaded with a signaling factor cocktail, obtained from secretome of hypoxia-conditioned human mesenchymal stem cells (MSC). Therefore, hypoxia-conditioned medium (HCM)-production is standardized and adapted to achieve high signaling factor-yields; a concentration protocol based on dialysis and freeze-drying is established to enable the integration of sufficient and defined amounts into the depot. In humid milieu-as after implantation-signaling factors are released by forming a chemotactic gradient, inducing a directed migration of human bone marrow stroma cells (hBMSC) into the scaffold. Angiogenic potential, determined by coculturing human umbilical vein endothelial cells (HUVEC) with osteogenically induced hBMSC shows prevascular structures, which sprout throughout the interconnected pores in a HCM-concentration-dependent manner. Retarded release by alginate-based (1 vol%) depots, significantly improves sprouting-depth and morphology of tubular structures. With the intrinsic potential to supply attracted cells with oxygen and nutrients, this bioactive material system has great potential for clinical translation.
Collapse
Affiliation(s)
- Mandy Quade
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Pina Münch
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Anja Lode
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Sarah Duin
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Corina Vater
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
- University Centre of Orthopaedica and TraumatologyFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Anastasia Gabrielyan
- Department of PediatricsFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Angela Rösen‐Wolff
- Department of PediatricsFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| | - Michael Gelinsky
- Centre for Translational BoneJoint and Soft Tissue ResearchFaculty of Medicine and University Hospital Carl Gustav CarusTechnische Universität Dresden Fetscherstr. 74 01307 Dresden Germany
| |
Collapse
|
31
|
Wang Z, Mithieux SM, Weiss AS. Fabrication Techniques for Vascular and Vascularized Tissue Engineering. Adv Healthc Mater 2019; 8:e1900742. [PMID: 31402593 DOI: 10.1002/adhm.201900742] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/12/2019] [Indexed: 12/19/2022]
Abstract
Impaired or damaged blood vessels can occur at all levels in the hierarchy of vascular systems from large vasculatures such as arteries and veins to meso- and microvasculatures such as arterioles, venules, and capillary networks. Vascular tissue engineering has become a promising approach for fabricating small-diameter vascular grafts for occlusive arteries. Vascularized tissue engineering aims to fabricate meso- and microvasculatures for the prevascularization of engineered tissues and organs. The ideal small-diameter vascular graft is biocompatible, bridgeable, and mechanically robust to maintain patency while promoting tissue remodeling. The desirable fabricated meso- and microvasculatures should rapidly integrate with the host blood vessels and allow nutrient and waste exchange throughout the construct after implantation. A number of techniques used, including engineering-based and cell-based approaches, to fabricate these synthetic vasculatures are herein explored, as well as the techniques developed to fabricate hierarchical structures that comprise multiple levels of vasculature.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Suzanne M. Mithieux
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
| | - Anthony S. Weiss
- School of Life and Environmental Sciences University of Sydney NSW 2006 Australia
- Charles Perkins Centre University of Sydney NSW 2006 Australia
- Bosch Institute University of Sydney NSW 2006 Australia
- Sydney Nano Institute University of Sydney NSW 2006 Australia
| |
Collapse
|
32
|
Sakthivel K, O'Brien A, Kim K, Hoorfar M. Microfluidic analysis of heterotypic cellular interactions: A review of techniques and applications. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.03.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
33
|
Charbe NB, Zacconi FC, Amnerkar N, Pardhi D, Shukla P, Mukattash TL, McCarron PA, Tambuwala MM. Emergence of Three Dimensional Printed Cardiac Tissue: Opportunities and Challenges in Cardiovascular Diseases. Curr Cardiol Rev 2019; 15:188-204. [PMID: 30648518 PMCID: PMC6719392 DOI: 10.2174/1573403x15666190112154710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 01/01/2023] Open
Abstract
Three-dimensional (3D) printing, also known as additive manufacturing, was developed originally for engineering applications. Since its early advancements, there has been a relentless de-velopment in enthusiasm for this innovation in biomedical research. It allows for the fabrication of structures with both complex geometries and heterogeneous material properties. Tissue engineering using 3D bio-printers can overcome the limitations of traditional tissue engineering methods. It can match the complexity and cellular microenvironment of human organs and tissues, which drives much of the interest in this technique. However, most of the preliminary evaluations of 3D-printed tissues and organ engineering, including cardiac tissue, relies extensively on the lessons learned from tradi-tional tissue engineering. In many early examples, the final printed structures were found to be no bet-ter than tissues developed using traditional tissue engineering methods. This highlights the fact that 3D bio-printing of human tissue is still very much in its infancy and more work needs to be done to realise its full potential. This can be achieved through interdisciplinary collaboration between engi-neers, biomaterial scientists and molecular cell biologists. This review highlights current advance-ments and future prospects for 3D bio-printing in engineering ex vivo cardiac tissue and associated vasculature, such as coronary arteries. In this context, the role of biomaterials for hydrogel matrices and choice of cells are discussed. 3D bio-printing has the potential to advance current research signif-icantly and support the development of novel therapeutics which can improve the therapeutic out-comes of patients suffering fatal cardiovascular pathologies.
Collapse
Affiliation(s)
- Nitin B Charbe
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna Mackenna 4860, Macul, Santiago 7820436, Chile.,Sri Adichunchunagiri College of Pharmacy, Sri Adichunchunagiri University, BG Nagar, Karnataka 571418, India
| | - Flavia C Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna Mackenna 4860, Macul, Santiago 7820436, Chile.,Institute of Biological and Medical Engineering, School of Engineering, Medicine and Biological Science, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nikhil Amnerkar
- Adv V. R. Manohar Institute of Diploma in Pharmacy, Wanadongri, Hingna Road, Nagpur, Maharashtra 441110, India
| | - Dinesh Pardhi
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zheijiang University, Hangzhou 310027, China
| | - Priyank Shukla
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Derry/Londonderry, BT47 6SB, Northern Ireland, United Kingdom
| | - Tareq L Mukattash
- Department of Clinical Pharmacy Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Paul A McCarron
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| |
Collapse
|
34
|
Xue D, Zhang J, Wang Y, Mei D. Digital Light Processing-Based 3D Printing of Cell-Seeding Hydrogel Scaffolds with Regionally Varied Stiffness. ACS Biomater Sci Eng 2019; 5:4825-4833. [DOI: 10.1021/acsbiomaterials.9b00696] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dai Xue
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiaxin Zhang
- Department of Toxicology, Fourth Military Medical University, Xi’an 710032, China
| | - Yancheng Wang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Deqing Mei
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of Advanced Manufacturing Technology of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
35
|
Heinrich MA, Liu W, Jimenez A, Yang J, Akpek A, Liu X, Pi Q, Mu X, Hu N, Schiffelers RM, Prakash J, Xie J, Zhang YS. 3D Bioprinting: from Benches to Translational Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1805510. [PMID: 31033203 PMCID: PMC6752725 DOI: 10.1002/smll.201805510] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/03/2019] [Indexed: 05/07/2023]
Abstract
Over the last decades, the fabrication of 3D tissues has become commonplace in tissue engineering and regenerative medicine. However, conventional 3D biofabrication techniques such as scaffolding, microengineering, and fiber and cell sheet engineering are limited in their capacity to fabricate complex tissue constructs with the required precision and controllability that is needed to replicate biologically relevant tissues. To this end, 3D bioprinting offers great versatility to fabricate biomimetic, volumetric tissues that are structurally and functionally relevant. It enables precise control of the composition, spatial distribution, and architecture of resulting constructs facilitating the recapitulation of the delicate shapes and structures of targeted organs and tissues. This Review systematically covers the history of bioprinting and the most recent advances in instrumentation and methods. It then focuses on the requirements for bioinks and cells to achieve optimal fabrication of biomimetic constructs. Next, emerging evolutions and future directions of bioprinting are discussed, such as freeform, high-resolution, multimaterial, and 4D bioprinting. Finally, the translational potential of bioprinting and bioprinted tissues of various categories are presented and the Review is concluded by exemplifying commercially available bioprinting platforms.
Collapse
Affiliation(s)
- Marcel Alexander Heinrich
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, Enschede 7500AE, The Netherlands
| | - Wanjun Liu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Key Laboratory of Textile Science and Technology, College of Textiles, Donghua University, Shanghai 201620, P.R. China
| | - Andrea Jimenez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Biomedical Engineering Laboratory, Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Nuevo León 64849, Mexico
| | - Jingzhou Yang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Center of Biomedical Materials 3D Printing, National Engineering Laboratory for Polymer Complex Structure Additive Manufacturing, Baoding 071000, P.R. China
| | - Ali Akpek
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Biomedical Engineering, Istanbul Yeni Yuzyil University, Istanbul 34010, Turkey
| | - Xiao Liu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, P.R. China
| | - Qingmeng Pi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Plastic and Reconstructive Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200129, P.R. China
| | - Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ning Hu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, P.R. China
| | - Raymond Michel Schiffelers
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, Enschede 7500AE, The Netherlands
| | - Jingwei Xie
- Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
36
|
Cohen S, Liu Q, Wright M, Garvin J, Rarick K, Harder D. High glucose conditioned neonatal astrocytes results in impaired mitogenic activity in cerebral microvessel endothelial cells in co-culture. Heliyon 2019; 5:e01795. [PMID: 31193586 PMCID: PMC6536426 DOI: 10.1016/j.heliyon.2019.e01795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/22/2019] [Accepted: 05/20/2019] [Indexed: 11/20/2022] Open
Abstract
Angiogenesis is a highly complex and coordinated process in the brain. Under normal conditions, it is a vital process in growth and development, but under adverse conditions such as diabetes mellitus, it can lead to severe pathology. Astrocytes are a key constituent of the neurovascular unit and contribute to cerebral function, not only bridging the gap between metabolic supplies from blood vessels to neurons, but also regulating angiogenesis. Astrocytes affect angiogenesis by secreting angiogenic factors such as vascular endothelial growth factor (VEGF) into its microenvironment and regulating mitogenic activity in cerebral microvessel endothelial cells (CMEC). We hypothesized that astrocytes conditioned in high glucose media would produce and secrete decreased VEGF which would lead to impaired proliferation, migration, and tube formation of CMEC in vitro. Using neonatal rat astrocytes, we used normal glucose (NG, 5.5mM) vs. high glucose (HG, 25mM) feeding media and measured VEGF message and protein levels as well as secreted VEGF. We co-cultured conditioned astrocytes with isolated rat CMEC and measured mitogenic activity of endothelial cells using BrdU assay, scratch recovery assay, and tube formation assay. HG astrocytes produced and secreted decreased VEGF protein and resulted in impaired mitogenic activity when co-cultured with CMEC as demonstrated by decreased BrdU uptake, decreased scratch recovery, and slower tube formation. Our study provides insight into gliovascular adaptations to increased glucose levels resulting in impaired cellular cross-talk between astrocytes and CMEC which could be one explanation for cerebral microangiopathy seen in diabetic conditions.
Collapse
Affiliation(s)
- Susan Cohen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
- Corresponding author.
| | - Qiuli Liu
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Jodi Garvin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kevin Rarick
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David Harder
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
37
|
Renggli K, Rousset N, Lohasz C, Nguyen OTP, Hierlemann A. Integrated Microphysiological Systems: Transferable Organ Models and Recirculating Flow. ADVANCED BIOSYSTEMS 2019; 3:e1900018. [PMID: 32627410 PMCID: PMC7610576 DOI: 10.1002/adbi.201900018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Studying and understanding of tissue and disease mechanisms largely depend on the availability of suitable and representative biological model systems. These model systems should be carefully engineered and faithfully reproduce the biological system of interest to understand physiological effects, pharmacokinetics, and toxicity to better identify new drug compounds. By relying on microfluidics, microphysiological systems (MPSs) enable the precise control of culturing conditions and connections of advanced in vitro 3D organ models that better reproduce in vivo environments. This review focuses on transferable in vitro organ models and integrated MPSs that host these transferable biological units and enable interactions between different tissue types. Interchangeable and transferrable in vitro organ models allow for independent quality control of the biological model before system assembly and building MPS assays on demand. Due to the complexity and different maturation times of individual in vitro tissues, off-chip production and quality control entail improved stability and reproducibility of the systems and results, which is important for large-scale adoption of the technology. Lastly, the technical and biological challenges and open issues for realizing and implementing integrated MPSs with transferable in vitro organ models are discussed.
Collapse
Affiliation(s)
- Kasper Renggli
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | | | | | | |
Collapse
|
38
|
Heinemann C, Heinemann S, Rößler S, Kruppke B, Wiesmann HP, Hanke T. Organically modified hydroxyapatite (ormoHAP) nanospheres stimulate the differentiation of osteoblast and osteoclast precursors: a co-culture study. ACTA ACUST UNITED AC 2019; 14:035015. [PMID: 30870824 DOI: 10.1088/1748-605x/ab0fad] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Isolated nanospheres consisting of organically modified hydroxyapatite (ormoHAP), prepared by an electric field-assisted ion double migration process, were embedded in foamed gelatin to form a composite scaffold. Degradation rates have been demonstrated to correlate with the crosslinking degree (40%, 80%) as well as with the mineral content of the scaffolds (0%, 20%, 40%). A human co-culture model of osteoblasts and osteoclasts, derived from bone marrow stromal cells and monocytes, respectively, without external addition of the factors RANKL and M-CSF, was run for up to 42 d in order to characterize the action of the ormoHAP-gelatin scaffolds on the co-culture. Examination was performed by quantitative biochemical methods (DNA, LDH, ALP, TRAP5b), gene expression analysis (ALP, BSP II, RANKL, IL-6, VTNR, CTSK, TRAP, OSCAR, CALCR) and confocal laser scanning microscopy (cell nuclei, actin, CD68, TRAP). Results confirm that ormoHAP embedded in the gelatin matrix enhanced TRAP 5b activity. As a feedback, ALP activity and gene expression of BSP II of osteoblasts increased. Finally, a sequence of cell cross-talk actions is suggested, which can explain the behavior of the formed vital co-culture and moreover the influence of the presence and concentration of ormoHAP.
Collapse
|
39
|
Pereira RC, Benelli R, Canciani B, Scaranari M, Daculsi G, Cancedda R, Gentili C. Beta-tricalcium phosphate ceramic triggers fast and robust bone formation by human mesenchymal stem cells. J Tissue Eng Regen Med 2019; 13:1007-1018. [PMID: 30811859 DOI: 10.1002/term.2848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022]
Abstract
Due to their osteoconductive and inductive properties, a variety of calcium phosphate (CaP) scaffolds are commonly used in orthopaedics as graft material to heal bone defects. In this study, we have used two CaP scaffolds with different hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP) ratios (MBCP®; 60/40 and MBCP+ ®; 20/80) to investigate their intrinsic capacity to favour human bone marrow stem cells (hBMSCs) osteogenic differentiation capacity. We report that MBCP+ ® showed in in vitro culture model a higher rate of calcium ion release in comparison with MBCP®. In two defined coculture systems, the hBMSC seeded onto MBCP+ ® presented an increased amount of VEGF secretion, resulting in an enhanced endothelial cell proliferation and capillary formation compared with hBMSC seeded onto MBCP®. When both ceramics combined with hBMSC were implanted in a nude mouse model, we observed a faster osteogenic differentiation and enhancement mature bone deposition sustained by the presence of a vast host vasculature within the MBCP+ ® ceramics. Bone formation was observed in samples highly positive to the activation of calcium sensing receptor protein (CaSr) on the surface of seeded hBMSC that also shown higher BMP-2 protein expression. With these data we provide valuable insights in the possible mechanisms of ossification and angiogenesis by hBMSC that we believe to be primed by calcium ions released from CaP scaffolds. Evidences could lead to an optimization of ceramic scaffolds to prime bone repair.
Collapse
Affiliation(s)
- Rui C Pereira
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Roberto Benelli
- Laboratory of Immunology, IRCCS AOU San Martino, Genoa, Italy
| | - Barbara Canciani
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Monica Scaranari
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Guy Daculsi
- INSERM LIOAD U791, Dental Faculty, Nantes University, Nantes, France
| | - Ranieri Cancedda
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Chiara Gentili
- Laboratory of Regenerative Medicine, Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
40
|
Piard C, Baker H, Kamalitdinov T, Fisher J. Bioprinted osteon-like scaffolds enhance in vivo neovascularization. Biofabrication 2019; 11:025013. [PMID: 30769337 PMCID: PMC7195919 DOI: 10.1088/1758-5090/ab078a] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone tissue engineers are facing a daunting challenge when attempting to fabricate bigger constructs intended for use in the treatment of large bone defects, which is the vascularization of the graft. Cell-based approaches and, in particular, the use of in vitro coculture of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) has been one of the most explored options. We present in this paper an alternative method to mimic the spatial pattern of HUVECs and hMSCs found in native osteons based on the use of extrusion-based 3D bioprinting (3DP). We developed a 3DP biphasic osteon-like scaffold, containing two separate osteogenic and vasculogenic cell populations encapsulated in a fibrin bioink in order to improve neovascularization. To this end, we optimized the fibrin bioink to improve the resolution of printed strands and ensure a reproducible printing process; the influence of printing parameters on extruded strand diameter and cell survival was also investigated. The mechanical strength of the construct was improved by co-printing the fibrin bioink along a supporting PCL carrier scaffold. Compressive mechanical testing showed improved mechanical properties with an average compressive modulus of 131 ± 23 MPa, which falls in the range of cortical bone. HUVEC and hMSC laden fibrin hydrogels were printed in osteon-like patterns and cultured in vitro. A significant increase in gene expression of angiogenic markers was observed for the biomimetic scaffolds. Finally, biphasic scaffolds were implanted subcutaneously in rats. Histological analysis of explanted scaffolds showed a significant increase in the number of blood vessels per area in the 3D printed osteon-like scaffolds. The utilization of these scaffolds in constructing biomimetic osteons for bone regeneration demonstrated a promising capacity to improve neovascularization of the construct. These results indicates that proper cell orientation and scaffold design could play a critical role in neovascularization.
Collapse
Affiliation(s)
- Charlotte Piard
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| | - Hannah Baker
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| | - Timur Kamalitdinov
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| | - John Fisher
- Fischell Department of Bioengineering, University of Maryland, 3121 A James Clark Hall, College Park,MD20742, United States of America
- Center for Engineering Complex Tissues, University of Maryland, 3121 AJames Clark Hall, College Park,MD20742, United States of America
| |
Collapse
|
41
|
Jia Z, Guo H, Xie H, Zhou J, Wang Y, Bao X, Huang Y, Chen F. Construction of Pedicled Smooth Muscle Tissues by Combining the Capsule Tissue and Cell Sheet Engineering. Cell Transplant 2019; 28:328-342. [PMID: 30712374 PMCID: PMC6425107 DOI: 10.1177/0963689718821682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The survival of engineered tissue requires the formation of its own capillary network, which can anastomose with the host vasculature after transplantation. Currently, while many strategies, such as modifying the scaffold material, adding endothelial cells, or angiogenic factors, have been researched, engineered tissue implanted in vivo cannot timely access to sufficient blood supply, leading to ischemic apoptosis or shrinkage. Constructing vascularized engineered tissue with its own axial vessels and subsequent pedicled transfer is promising to solve the problem of vascularization in tissue engineering. In this study, we used the tissue expander capsule as a novel platform for vascularizing autologous smooth muscle cell (SMC) sheets and fabricating vascularized engineered tissue with its own vascular pedicle. First, we verified which time point was the most effective for constructing an axial capsule vascular bed. Second, we compared the outcome of SMC sheet transplantation onto the expander capsule and classical dorsal subcutaneous tissue, which was widely used in other studies for vascularization. Finally, we transplanted multilayered SMC sheets onto the capsule bed twice to verify the feasibility of fabricating thick pedicled engineered smooth muscle tissues. The results indicated that the axial capsule tissue could be successfully induced, and the capsule tissue 1 week after full expansion was the most vascularized. Quantitative comparisons of thickness, vessel density, and apoptosis of cell sheet grafts onto two vascular beds proved that the axial capsule vascular bed was more favorable to the growth and vascularization of transplants than classical subcutaneous tissue. Furthermore, thick vascularized smooth muscle tissues with the vascular pedicle could be constructed by multi-transplanting cell sheets onto the capsule bed. The combination of axial capsule vascular bed and cell sheet engineering may provide an efficient strategy to overcome the problem of slow or insufficient vascularization in tissue engineering.
Collapse
Affiliation(s)
- Zhiming Jia
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hailin Guo
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Xie
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Junmei Zhou
- 2 Department of Central Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaping Wang
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xingqi Bao
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yichen Huang
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fang Chen
- 1 Department of Urology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Gariboldi MI, Butler R, Best SM, Cameron RE. Engineering vasculature: Architectural effects on microcapillary-like structure self-assembly. PLoS One 2019; 14:e0210390. [PMID: 30620757 PMCID: PMC6324810 DOI: 10.1371/journal.pone.0210390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/21/2018] [Indexed: 01/24/2023] Open
Abstract
One of the greatest obstacles to clinical translation of bone tissue engineering is the inability to effectively and efficiently vascularize scaffolds. The goal of this work was to explore systematically whether architecture, at a scale of hundreds of microns, can be used to direct the growth of microcapillary-like structures into the core of scaffolds. Biphasic bioceramic patterned architectures were produced using silicone molds of 3D printed parts. Grooves and ridges were designed to have widths of 330 μm and 660 μm, with periodicities respectively of 1240 μm and 630 μm. Groove depth was varied between 150 μm and 585 μm. Co-cultures of human dermal microvascular endothelial cells (HDMECs) and human osteoblasts (hOBs) were used to grow microcapillary-like structures on substrates. Bioceramic architecture was found to significantly affect microcapillary-like structure location and orientation. Microcapillary-like structures were found to form predominantly in grooves or between convexities. For all patterned samples, the CD31 (endothelial cell marker) signal was at least 2.5 times higher along grooves versus perpendicular to grooves. In addition, the average signal was at least two times higher within grooves than outside grooves for all samples. Grooves with a width of 330 μm and a depth of 300 μm resulted in the formation of individual, highly aligned microcapillary-like structures with lengths around 5 mm. Extensive literature has focused on the role of nano- and micro-topography (on the scale below tens of microns) on cellular response. However, the idea that architecture at a scale much larger than a cell could be used to modulate angiogenesis has not been systematically investigated. This work shows the crucial influence of architecture on microcapillary-like structure self-assembly at the scale of hundreds of microns. Elucidating the precise correspondence between architecture and microcapillary-like structure organization will ultimately allow the engineering of microvasculature by tuning local scaffold design to achieve desirable microvessel properties.
Collapse
Affiliation(s)
- Maria Isabella Gariboldi
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, University of Cambridge, Cambridge, United Kingdom
| | - Richard Butler
- Imaging Facility, Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Serena M. Best
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, University of Cambridge, Cambridge, United Kingdom
| | - Ruth E. Cameron
- Cambridge Centre for Medical Materials, Department of Materials Science and Metallurgy, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
43
|
Torras N, García-Díaz M, Fernández-Majada V, Martínez E. Mimicking Epithelial Tissues in Three-Dimensional Cell Culture Models. Front Bioeng Biotechnol 2018; 6:197. [PMID: 30619844 PMCID: PMC6305315 DOI: 10.3389/fbioe.2018.00197] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
Epithelial tissues are composed of layers of tightly connected cells shaped into complex three-dimensional (3D) structures such as cysts, tubules, or invaginations. These complex 3D structures are important for organ-specific functions and often create biochemical gradients that guide cell positioning and compartmentalization within the organ. One of the main functions of epithelia is to act as physical barriers that protect the underlying tissues from external insults. In vitro, epithelial barriers are usually mimicked by oversimplified models based on cell lines grown as monolayers on flat surfaces. While useful to answer certain questions, these models cannot fully capture the in vivo organ physiology and often yield poor predictions. In order to progress further in basic and translational research, disease modeling, drug discovery, and regenerative medicine, it is essential to advance the development of new in vitro predictive models of epithelial tissues that are capable of representing the in vivo-like structures and organ functionality more accurately. Here, we review current strategies for obtaining biomimetic systems in the form of advanced in vitro models that allow for more reliable and safer preclinical tests. The current state of the art and potential applications of self-organized cell-based systems, organ-on-a-chip devices that incorporate sensors and monitoring capabilities, as well as microfabrication techniques including bioprinting and photolithography, are discussed. These techniques could be combined to help provide highly predictive drug tests for patient-specific conditions in the near future.
Collapse
Affiliation(s)
- Núria Torras
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - María García-Díaz
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Vanesa Fernández-Majada
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Elena Martínez
- Biomimetic Systems for Cell Engineering, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red, Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
44
|
Weber D, Knaak S, Hettrich K, Andrulis M, Momburg F, Quade M, Gelinsky M, Schwartz-Albiez R. Influence of Regioselectively Sulfated Cellulose on in Vitro Vascularization of Biomimetic Bone Matrices. Biomacromolecules 2018; 19:4228-4238. [DOI: 10.1021/acs.biomac.8b01004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dominik Weber
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Immunology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sven Knaak
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscher Strasse 74, 1307 Dresden, Germany
| | - Kay Hettrich
- Fraunhofer Institut für Angewandte Polymerforschung (IAP), Geiselbergstrasse 69, 14476 Potsdam-Golm Germany
| | - Mindaugas Andrulis
- Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Frank Momburg
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Immunology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Mandy Quade
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscher Strasse 74, 1307 Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscher Strasse 74, 1307 Dresden, Germany
| | - Reinhard Schwartz-Albiez
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Applied Tumor Immunology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
45
|
Skrzypek K, Nibbelink MG, Karbaat LP, Karperien M, van Apeldoorn A, Stamatialis D. An important step towards a prevascularized islet macroencapsulation device-effect of micropatterned membranes on development of endothelial cell network. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:91. [PMID: 29938334 PMCID: PMC6018599 DOI: 10.1007/s10856-018-6102-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/05/2018] [Indexed: 05/23/2023]
Abstract
The development of immune protective islet encapsulation devices could allow for islet transplantation in the absence of immunosuppression. However, the immune protective membrane / barrier introduced there could also impose limitations in transport of oxygen and nutrients to the encapsulated cells resulting to limited islet viability. In the last years, it is well understood that achieving prevascularization of the device in vitro could facilitate its connection to the host vasculature after implantation, and therefore could provide sufficient blood supply and oxygenation to the encapsulated islets. However, the microvascular networks created in vitro need to mimic well the highly organized vasculature of the native tissue. In earlier study, we developed a functional macroencapsulation device consisting of two polyethersulfone/polyvinylpyrrolidone (PES/PVP) membranes, where a bottom microwell membrane provides good separation of encapsulated islets and the top flat membrane acts as a lid. In this work, we investigate the possibility of creating early microvascular networks on the lid of this device by combining novel membrane microfabrication with co-culture of human umbilical vein endothelial cell (HUVEC) and fibroblasts. We create thin porous microstructured PES/PVP membranes with solid and intermittent line-patterns and investigate the effect of cell alignment and cell interconnectivity as a first step towards the development of a stable prevascularized layer in vitro. Our results show that, in contrast to non-patterned membranes where HUVECs form unorganized HUVEC branch-like structures, for the micropatterned membranes, we can achieve cell alignment and the co-culture of HUVECs on a monolayer of fibroblasts attached on the membranes with intermittent line-pattern allows for the creation of HUVEC branch-like structures over the membrane surface. This important step towards creating early microvascular networks was achieved without the addition of hydrogels, often used in angiogenesis assays, as gels could block the pores of the membrane and limit the transport properties of the islet encapsulation device.
Collapse
Affiliation(s)
- Katarzyna Skrzypek
- Bioartificial organs, Biomaterials Science and Technology, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - Milou Groot Nibbelink
- Developmental BioEngineering, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Lisanne P Karbaat
- Developmental BioEngineering, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Marcel Karperien
- Developmental BioEngineering, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Aart van Apeldoorn
- Developmental BioEngineering, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Complex Tissue Regeneration, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dimitrios Stamatialis
- Bioartificial organs, Biomaterials Science and Technology, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| |
Collapse
|
46
|
Xue D, Wang Y, Zhang J, Mei D, Wang Y, Chen S. Projection-Based 3D Printing of Cell Patterning Scaffolds with Multiscale Channels. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19428-19435. [PMID: 29782142 DOI: 10.1021/acsami.8b03867] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To fully actualize artificial, cell-laden biological models in tissue engineering, such as 3D organoids and organs-on-a-chip systems, cells need to be patterned such that they can precisely mimic natural microenvironments in vitro. Despite increasing interest in this area, patterning cells at multiscale (∼10 μm to 10 mm) remains a significant challenge in bioengineering. Here, we report a projection-based 3D printing system that achieves rapid and high-resolution fabrication of hydrogel scaffolds featuring intricate channels for multiscale cell patterning. Using this system, we were able to use biocompatible poly(ethylene glycol)diacrylate in fabricating a variety of scaffold architectures, ranging from regular geometries such as serpentine, spiral, and fractal-like to more irregular/intricate geometries, such as biomimetic arborescent and capillary networks. A red food dye solution was able to freely fill all channels in the scaffolds, from the trunk (>1100 μm in width) to the small branch (∼17 μm in width) without an external pump. The dimensions of the printed scaffolds remained stable over 3 days while being immersed in Dulbecco's phosphate-buffered saline at 37 °C, and a penetration analysis revealed that these scaffolds are suitable for metabolic and nutrient transport. Cell patterning experiments showed that red fluorescent protein-transfected A549 human nonsmall lung cancer cells adhered well in the scaffolds' channels, and showed further attachment and penetration during cell culture proliferation.
Collapse
Affiliation(s)
- Dai Xue
- Department of NanoEngineering , University of California , San Diego , California 92093 , United States
| | | | - Jiaxin Zhang
- Department of Toxicology , Fourth Military Medical University , Xi'an 710032 , China
| | | | | | - Shaochen Chen
- Department of NanoEngineering , University of California , San Diego , California 92093 , United States
| |
Collapse
|
47
|
Chai YC, Mendes LF, van Gastel N, Carmeliet G, Luyten FP. Fine-tuning pro-angiogenic effects of cobalt for simultaneous enhancement of vascular endothelial growth factor secretion and implant neovascularization. Acta Biomater 2018; 72:447-460. [PMID: 29626696 DOI: 10.1016/j.actbio.2018.03.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/25/2018] [Accepted: 03/28/2018] [Indexed: 02/07/2023]
Abstract
Rapid neovascularization of a tissue-engineered (TE) construct by the host vasculature is quintessential to warrant effective bone regeneration. This process can be promoted through active induction of angiogenic growth factor secretion or by implementation of in vitro pre-vascularization strategies. In this study, we aimed at optimizing the pro-angiogenic effect of Cobalt (Co2+) to enhance vascular endothelial growth factor (VEGF) expression by human periosteum-derived mesenchymal stem cells (hPDCs). Simultaneously we set out to promote microvascular network formation by co-culturing with human umbilical vein endothelial cells (HUVECs). The results showed that Co2+ treatments (at 50, 100 or 150 µM) significantly upregulated in vitro VEGF expression, but inhibited hPDCs growth and HUVECs network formation in co-cultures. These inhibitory effects were mitigated at lower Co2+ concentrations (at 5, 10 or 25 µM) while VEGF expression remained significantly upregulated and further augmented in the presence of Ascorbic Acid and Dexamethasone possibly through Runx2 upregulation. The supplements also facilitated HUVECs network formation, which was dependent on the quantity and spatial distribution of collagen type-1 matrix deposited by the hPDCs. When applied to hPDCs seeded onto calcium phosphate scaffolds, the supplements significantly induced VEGF secretion in vitro, and promoted higher vascularization upon ectopic implantation in nude mice shown by an increase of CD31 positive blood vessels within the scaffolds. Our findings provided novel insights into the pleotropic effects of Co2+ on angiogenesis (i.e. promoted VEGF secretion and inhibited endothelial network formation), and showed potential to pre-condition TE constructs under one culture regime for improved implant neovascularization in vivo. STATEMENT OF SIGNIFICANT Cobalt (Co2+) is known to upregulate vascular endothelial growth factor (VEGF) secretion, however it also inhibits in vitro angiogenesis through unknown Co2+-induced events. This limits the potential of Co2+ for pro-angiogenesis of tissue engineered (TE) implants. We showed that Co2+ upregulated VEGF expression by human periosteum-derived cells (hPDCs) but reduced the cell growth, and endothelial network formation due to reduction of col-1 matrix deposition. Supplementation with Ascorbic acid and Dexamethasone concurrently improved hPDCs growth, endothelial network formation, and upregulated VEGF secretion. In vitro pre-conditioning of hPDC-seeded TE constructs with this fine-tuned medium enhanced VEGF secretion and implant neovascularization. Our study provided novel insights into the pleotropic effects of Co2+ on angiogenesis and formed the basis for improving implant neovascularization.
Collapse
|
48
|
Yuan Q, Arkudas A, Horch RE, Hammon M, Bleiziffer O, Uder M, Seuss H. Vascularization of the Arteriovenous Loop in a Rat Isolation Chamber Model—Quantification of Hypoxia and Evaluation of Its Effects. Tissue Eng Part A 2018; 24:719-728. [DOI: 10.1089/ten.tea.2017.0262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Quan Yuan
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Matthias Hammon
- Department of Radiology, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Oliver Bleiziffer
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
- Department of Plastic and Hand Surgery, Inselspital Bern, Universität Bern, Bern, Switzerland
| | - Michael Uder
- Department of Radiology, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| | - Hannes Seuss
- Department of Radiology, University Hospital Erlangen, Friedrich Alexander University, Erlangen-Nuernberg (FAU), Erlangen, Germany
| |
Collapse
|
49
|
Kargozar S, Baino F, Hamzehlou S, Hill RG, Mozafari M. Bioactive Glasses: Sprouting Angiogenesis in Tissue Engineering. Trends Biotechnol 2018; 36:430-444. [DOI: 10.1016/j.tibtech.2017.12.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 02/08/2023]
|
50
|
Heher P, Mühleder S, Mittermayr R, Redl H, Slezak P. Fibrin-based delivery strategies for acute and chronic wound healing. Adv Drug Deliv Rev 2018; 129:134-147. [PMID: 29247766 DOI: 10.1016/j.addr.2017.12.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/24/2017] [Accepted: 12/09/2017] [Indexed: 12/17/2022]
Abstract
Fibrin, a natural hydrogel, is the end product of the physiological blood coagulation cascade and naturally involved in wound healing. Beyond its role in hemostasis, it acts as a local reservoir for growth factors and as a provisional matrix for invading cells that drive the regenerative process. Its unique intrinsic features do not only promote wound healing directly via modulation of cell behavior but it can also be fine-tuned to evolve into a delivery system for sustained release of therapeutic biomolecules, cells and gene vectors. To further augment tissue regeneration potential, current strategies exploit and modify the chemical and physical characteristics of fibrin to employ combined incorporation of several factors and their timed release. In this work we show advanced therapeutic approaches employing fibrin matrices in wound healing and cover the many possibilities fibrin offers to the field of regenerative medicine.
Collapse
|