1
|
Landau S, Kieda J, Khosravi R, Okhovatian S, Ramsay K, Liu C, Shakeri A, Zhao Y, Shen K, Bar-Am O, Levenberg S, Tsai S, Radisic M. Cell driven elastomeric particle packing in composite bioinks for engineering and implantation of stable 3D printed structures. Bioact Mater 2025; 44:411-427. [PMID: 39525804 PMCID: PMC11550138 DOI: 10.1016/j.bioactmat.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Geometric and structural integrity often deteriorate in 3D printed cell-laden constructs over time due to cellular compaction and hydrogel shrinkage. This study introduces a new approach that synergizes the advantages of cell compatibility of biological hydrogels and mechanical stability of elastomeric polymers for structure fidelity maintenance upon stereolithography and extrusion 3D printing. Enabling this advance is the composite bioink, formulated by integrating elastomeric microparticles from poly(octamethylene maleate (anhydride) citrate) (POMaC) into biologically derived hydrogels (fibrin, gelatin methacryloyl (GelMA), and alginate). The composite bioink enhanced the elasticity and plasticity of the 3D printed constructs, effectively mitigating tissue compaction and swelling. It exhibited a low shear modulus and a rapid crosslinking time, along with a high ultimate compressive strength and resistance to deformation from cellular forces and physical handling; this was attributed to packing and stress dissipation of elastomeric particles, which was confirmed via mathematical modelling. Enhanced functional assembly and stability of human iPSC-derived cardiac tissues and primary vasculature proved the utility of the composite bioink in tissue engineering. In vivo implantation studies revealed that constructs containing POMaC particles exhibited improved resilience against host tissue stress, enhanced angiogenesis, and infiltration of pro-reparative macrophages.
Collapse
Affiliation(s)
- Shira Landau
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Jennifer Kieda
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Ramak Khosravi
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Sargol Okhovatian
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Kaitlyn Ramsay
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Chuan Liu
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Amid Shakeri
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Yimu Zhao
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
- Acceleration Consortium, University of Toronto, Toronto, ON, Canada
| | - Karen Shen
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Orit Bar-Am
- Faculty of Biomedical Engineering, Technion, Haifa, IL, Israel
| | | | - Scott Tsai
- Toronto Metropolitan University, Department of Mechanical, Industrial, and Mechatronics Engineering, Toronto, ON, Canada
- Toronto Metropolitan University and Unity Health Toronto, Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, ON, Canada
| | - Milica Radisic
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
- University of Toronto, Department of Chemical Engineering and Applied Chemistry, Toronto, ON, Canada
- Acceleration Consortium, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Fang L, Lin X, Xu R, Liu L, Zhang Y, Tian F, Li JJ, Xue J. Advances in the Development of Gradient Scaffolds Made of Nano-Micromaterials for Musculoskeletal Tissue Regeneration. NANO-MICRO LETTERS 2024; 17:75. [PMID: 39601962 PMCID: PMC11602939 DOI: 10.1007/s40820-024-01581-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
The intricate hierarchical structure of musculoskeletal tissues, including bone and interface tissues, necessitates the use of complex scaffold designs and material structures to serve as tissue-engineered substitutes. This has led to growing interest in the development of gradient bone scaffolds with hierarchical structures mimicking the extracellular matrix of native tissues to achieve improved therapeutic outcomes. Building on the anatomical characteristics of bone and interfacial tissues, this review provides a summary of current strategies used to design and fabricate biomimetic gradient scaffolds for repairing musculoskeletal tissues, specifically focusing on methods used to construct compositional and structural gradients within the scaffolds. The latest applications of gradient scaffolds for the regeneration of bone, osteochondral, and tendon-to-bone interfaces are presented. Furthermore, the current progress of testing gradient scaffolds in physiologically relevant animal models of skeletal repair is discussed, as well as the challenges and prospects of moving these scaffolds into clinical application for treating musculoskeletal injuries.
Collapse
Affiliation(s)
- Lei Fang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xiaoqi Lin
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Ruian Xu
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yu Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Feng Tian
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
3
|
Wang Y, Han J, Tang W, Zhang X, Ding J, Xu Z, Song W, Li X, Wang L. Revealing transport, uptake and damage of polystyrene microplastics using a gut-liver-on-a-chip. LAB ON A CHIP 2024. [PMID: 39589486 DOI: 10.1039/d4lc00578c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Microplastics (MPs) are pervasive pollutants present in various environments. They have the capability to infiltrate the human gastrointestinal tract through avenues like water and food, and ultimately accumulating within the liver. However, due to the absence of reliable platforms, the transportation, uptake, and damage of microplastics in the gut-liver axis remain unclear. Here, we present the development of a gut-liver-on-a-chip (GLOC) featuring biomimetic intestinal peristalsis and a dynamic hepatic flow environment, exploring the translocation in the intestines and accumulation in the liver of MPs following oral ingestion. In comparison to conventional co-culture platforms, this chip has the capability to mimic essential physical microenvironments found within the intestines and liver (e.g., intestinal peristalsis and liver blood flow). It effectively reproduces the physiological characteristics of the intestine and liver (e.g., intestinal barrier and liver metabolism). Moreover, we infused polyethylene MPs with a diameter of 100 nm into the intestinal and hepatic chambers (concentrations ranging from 0 to 1 mg mL-1). We observed that as intestinal peristalsis increased (0%, 1%, 3%, 5%), the transport rate of MPs decreased, while the levels of oxidative stress and damage in hepatic cells decreased correspondingly. Our GLOC elucidates the process of MP transport in the intestine and uptake in the liver following oral ingestion. It underscores the critical role of intestinal peristalsis in protecting the liver from damage, and provides a novel research platform for assessing the organ-specific effects of MPs.
Collapse
Affiliation(s)
- Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenteng Tang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Xiaolong Zhang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Jiemeng Ding
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Zhipeng Xu
- Division of Clinical Medicine School of Medicine & Population Health University of Sheffield Medical School Beech Hill Road, Sheffield S10 2RX, UK
| | - Wei Song
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.
| | - Xinyu Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
4
|
Lei Q, Jia J, Guan X, Han K, Liu J, Duan R, Lian X, Huang D. Electrohydrodynamic Printing of Microscale Fibrous Scaffolds with a Sinusoidal Structure for Enhancing the Contractility of Cardiomyocytes. ACS Biomater Sci Eng 2024; 10:7227-7234. [PMID: 39390708 DOI: 10.1021/acsbiomaterials.4c00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mimicking the curved collagenous fibers in the cardiac extracellular matrix to fabricate elastic scaffolds in vitro is important for cardiac tissue engineering. Here, we developed sinusoidal polycaprolactone (PCL) fibrous scaffolds with commendable flexibility and elasticity to enhance the contractility of primary cardiomyocytes by employing melt-based electrohydrodynamic (EHD) printing. Microscale sinusoidal PCL fibers with an average diameter of ∼10 μm were printed to mimic the collagenous fibers in the cardiac ECM. The sinusoidal PCL fibrous scaffolds were EHD-printed in a layer-by-layer manner and exhibited outstanding flexibility and elasticity compared with the straight ones. The sinusoidal PCL scaffolds provided an elastic microenvironment for the attaching and spreading of primary cardiomyocytes, which facilitated their synchronous contractive activities. Primary cardiomyocytes also showed improved gene expression and maturation on the sinusoidal PCL scaffolds under electrical stimulation for 5 days. It is envisioned that the proposed flexible fibrous scaffold with biomimetic architecture may serve as a suitable patch for tissue regeneration and repair of damaged hearts after myocardial infarction.
Collapse
Affiliation(s)
- Qi Lei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China
| | - Jinqiao Jia
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Xiaomin Guan
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Kang Han
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Junzheng Liu
- Department of Bone and Joint Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, PR China
| | - Ruxin Duan
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China
| | - Xiaojie Lian
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China
| |
Collapse
|
5
|
Mirzajani H, Kraft M. Soft Bioelectronics for Heart Monitoring. ACS Sens 2024; 9:4328-4363. [PMID: 39239948 DOI: 10.1021/acssensors.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Cardiovascular diseases (CVDs) are a predominant global health concern, accounting for over 17.9 million deaths in 2019, representing approximately 32% of all global fatalities. In North America and Europe, over a million adults undergo cardiac surgeries annually. Despite the benefits, such surgeries pose risks and require precise postsurgery monitoring. However, during the postdischarge period, where monitoring infrastructures are limited, continuous monitoring of vital signals is hindered. In this area, the introduction of implantable electronics is altering medical practices by enabling real-time and out-of-hospital monitoring of physiological signals and biological information postsurgery. The multimodal implantable bioelectronic platforms have the capability of continuous heart sensing and stimulation, in both postsurgery and out-of-hospital settings. Furthermore, with the emergence of machine learning algorithms into healthcare devices, next-generation implantables will benefit artificial intelligence (AI) and connectivity with skin-interfaced electronics to provide more precise and user-specific results. This Review outlines recent advancements in implantable bioelectronics and their utilization in cardiovascular health monitoring, highlighting their transformative deployment in sensing and stimulation to the heart toward reaching truly personalized healthcare platforms compatible with the Sustainable Development Goal 3.4 of the WHO 2030 observatory roadmap. This Review also discusses the challenges and future prospects of these devices.
Collapse
Affiliation(s)
- Hadi Mirzajani
- Department of Electrical and Electronics Engineering, Koç University, Rumelifeneri Yolu, Sarıyer, Istanbul, 34450 Turkey
| | - Michael Kraft
- Department of Electrical Engineering (ESAT-MNS), KU Leuven, 3000 Leuven, Belgium
- Leuven Institute for Micro- and Nanoscale Integration (LIMNI), KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
6
|
Popovic AM, Lei MHC, Shakeri A, Khosravi R, Radisic M. Lab-on-a-chip models of cardiac inflammation. BIOMICROFLUIDICS 2024; 18:051507. [PMID: 39483204 PMCID: PMC11524635 DOI: 10.1063/5.0231735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024]
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide with numerous inflammatory cell etiologies associated with impaired cardiac function and heart failure. Inflammatory cardiomyopathy, also known as myocarditis, is an acquired cardiomyopathy characterized by inflammatory cell infiltration into the myocardium with a high risk of progression to deteriorated cardiac function. Recently, amidst the ongoing COVID-19 pandemic, the emergence of acute myocarditis as a complication of SARS-CoV-2 has garnered significant concern. Given its mechanisms remain elusive in conjunction with the recent withdrawal of previously FDA-approved antiviral therapeutics and prophylactics due to unexpected cardiotoxicity, there is a pressing need for human-mimetic platforms to investigate disease pathogenesis, model dysfunctional features, and support pre-clinical drug screening. Traditional in vitro models for studying cardiovascular diseases have inherent limitations in recapitulating the complexity of the in vivo microenvironment. Heart-on-a-chip technologies, combining microfabrication, microfluidics, and tissue engineering techniques, have emerged as a promising approach for modeling inflammatory cardiac diseases like myocarditis. This review outlines the established and emerging conditions of inflamed myocardium, identifying key features essential for recapitulating inflamed myocardial structure and functions in heart-on-a-chip models, highlighting recent advancements, including the integration of anisotropic contractile geometry, cardiomyocyte maturity, electromechanical functions, vascularization, circulating immunity, and patient/sex specificity. Finally, we discuss the limitations and future perspectives necessary for the clinical translation of these advanced technologies.
Collapse
Affiliation(s)
- Anna Maria Popovic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Matthew Ho Cheong Lei
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | | | | |
Collapse
|
7
|
Li X, Huo R, Li L, Cherif H, Lan X, Weber MH, Haglund L, Li J. Composite Hydrogel Sealants for Annulus Fibrosus Repair. ACS Biomater Sci Eng 2024; 10:5094-5107. [PMID: 38979636 DOI: 10.1021/acsbiomaterials.4c00548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Intervertebral disc (IVD) herniation is a leading cause of disability and lower back pain, causing enormous socioeconomic burdens. The standard of care for disc herniation is nucleotomy, which alleviates pain but does not repair the annulus fibrosus (AF) defect nor recover the biomechanical function of the disc. Existing bioadhesives for AF repair are limited by insufficient adhesion and significant mechanical and geometrical mismatch with the AF tissue, resulting in the recurrence of protrusion or detachment of bioadhesives. Here, we report a composite hydrogel sealant constructed from a composite of a three-dimensional (3D)-printed thermoplastic polyurethane (TPU) mesh and tough hydrogel. We tailored the fiber angle and volume fraction of the TPU mesh design to match the angle-ply structure and mechanical properties of native AF. Also, we proposed and tested three types of geometrical design of the composite hydrogel sealant to match the defect shape and size. Our results show that the sealant could mimic native AF in terms of the elastic modulus, flexural modulus, and fracture toughness and form strong adhesion with the human AF tissue. The bovine IVD tests show the effectiveness of the composite hydrogel sealant for AF repair and biomechanics recovery and for preventing herniation with its heightened stiffness and superior adhesion. By harnessing the combined capabilities of 3D printing and bioadhesives, these composite hydrogel sealants demonstrate promising potential for diverse applications in tissue repair and regeneration.
Collapse
Affiliation(s)
- Xuan Li
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec H3A 0C3, Canada
| | - Ran Huo
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec H3A 0C3, Canada
| | - Li Li
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G 1A3, Canada
| | - Hosni Cherif
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G 1A3, Canada
| | - Xiaoyi Lan
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec H3A 0C3, Canada
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G 1A3, Canada
| | - Michael H Weber
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G 1A3, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G 1A3, Canada
- Shriners Hospital for Children, 1003 Decarie Blvd, Montreal, Montreal, Quebec H4A 0A9, Canada
| | - Jianyu Li
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec H3A 0C3, Canada
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G 1A3, Canada
- Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
8
|
Janićijević Ž, Huang T, Bojórquez DIS, Tonmoy TH, Pané S, Makarov D, Baraban L. Design and Development of Transient Sensing Devices for Healthcare Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307232. [PMID: 38484201 PMCID: PMC11132064 DOI: 10.1002/advs.202307232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/12/2023] [Indexed: 05/29/2024]
Abstract
With the ever-growing requirements in the healthcare sector aimed at personalized diagnostics and treatment, continuous and real-time monitoring of relevant parameters is gaining significant traction. In many applications, health status monitoring may be carried out by dedicated wearable or implantable sensing devices only within a defined period and followed by sensor removal without additional risks for the patient. At the same time, disposal of the increasing number of conventional portable electronic devices with short life cycles raises serious environmental concerns due to the dangerous accumulation of electronic and chemical waste. An attractive solution to address these complex and contradictory demands is offered by biodegradable sensing devices. Such devices may be able to perform required tests within a programmed period and then disappear by safe resorption in the body or harmless degradation in the environment. This work critically assesses the design and development concepts related to biodegradable and bioresorbable sensors for healthcare applications. Different aspects are comprehensively addressed, from fundamental material properties and sensing principles to application-tailored designs, fabrication techniques, and device implementations. The emerging approaches spanning the last 5 years are emphasized and a broad insight into the most important challenges and future perspectives of biodegradable sensors in healthcare are provided.
Collapse
Affiliation(s)
- Željko Janićijević
- Institute of Radiopharmaceutical Cancer ResearchHelmholtz‐Zentrum Dresden‐Rossendorf e. V.01328DresdenGermany
| | - Tao Huang
- Institute of Radiopharmaceutical Cancer ResearchHelmholtz‐Zentrum Dresden‐Rossendorf e. V.01328DresdenGermany
| | | | - Taufhik Hossain Tonmoy
- Institute of Radiopharmaceutical Cancer ResearchHelmholtz‐Zentrum Dresden‐Rossendorf e. V.01328DresdenGermany
| | - Salvador Pané
- Multi‐Scale Robotics Lab (MSRL)Institute of Robotics & Intelligent Systems (IRIS)ETH ZürichZürich8092Switzerland
| | - Denys Makarov
- Institute of Ion Beam Physics and Materials ResearchHelmholtz‐Zentrum Dresden‐Rossendorf e. V.01328DresdenGermany
| | - Larysa Baraban
- Institute of Radiopharmaceutical Cancer ResearchHelmholtz‐Zentrum Dresden‐Rossendorf e. V.01328DresdenGermany
| |
Collapse
|
9
|
Kieda J, Shakeri A, Landau S, Wang EY, Zhao Y, Lai BF, Okhovatian S, Wang Y, Jiang R, Radisic M. Advances in cardiac tissue engineering and heart-on-a-chip. J Biomed Mater Res A 2024; 112:492-511. [PMID: 37909362 PMCID: PMC11213712 DOI: 10.1002/jbm.a.37633] [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: 07/05/2023] [Revised: 09/26/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
Recent advances in both cardiac tissue engineering and hearts-on-a-chip are grounded in new biomaterial development as well as the employment of innovative fabrication techniques that enable precise control of the mechanical, electrical, and structural properties of the cardiac tissues being modelled. The elongated structure of cardiomyocytes requires tuning of substrate properties and application of biophysical stimuli to drive its mature phenotype. Landmark advances have already been achieved with induced pluripotent stem cell-derived cardiac patches that advanced to human testing. Heart-on-a-chip platforms are now commonly used by a number of pharmaceutical and biotechnology companies. Here, we provide an overview of cardiac physiology in order to better define the requirements for functional tissue recapitulation. We then discuss the biomaterials most commonly used in both cardiac tissue engineering and heart-on-a-chip, followed by the discussion of recent representative studies in both fields. We outline significant challenges common to both fields, specifically: scalable tissue fabrication and platform standardization, improving cellular fidelity through effective tissue vascularization, achieving adult tissue maturation, and ultimately developing cryopreservation protocols so that the tissues are available off the shelf.
Collapse
Affiliation(s)
- Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Benjamin Fook Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Karamzadeh V, Shen ML, Ravanbakhsh H, Sohrabi‐Kashani A, Okhovatian S, Savoji H, Radisic M, Juncker D. High-Resolution Additive Manufacturing of a Biodegradable Elastomer with A Low-Cost LCD 3D Printer. Adv Healthc Mater 2024; 13:e2303708. [PMID: 37990819 PMCID: PMC11468968 DOI: 10.1002/adhm.202303708] [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: 10/25/2023] [Revised: 11/11/2023] [Indexed: 11/23/2023]
Abstract
Artificial organs and organs-on-a-chip (OoC) are of great clinical and scientific interest and have recently been made by additive manufacturing, but depend on, and benefit from, biocompatible, biodegradable, and soft materials. Poly(octamethylene maleate (anhydride) citrate (POMaC) meets these criteria and has gained popularity, and as in principle, it can be photocured and is amenable to vat-photopolymerization (VP) 3D printing, but only low-resolution structures have been produced so far. Here, a VP-POMaC ink is introduced and 3D printing of 80 µm positive features and complex 3D structures is demonstrated using low-cost (≈US$300) liquid-crystal display (LCD) printers. The ink includes POMaC, a diluent and porogen additive to reduce viscosity within the range of VP, and a crosslinker to speed up reaction kinetics. The mechanical properties of the cured ink are tuned to match the elastic moduli of different tissues simply by varying the porogen concentration. The biocompatibility is assessed by cell culture which yielded 80% viability and the potential for tissue engineering illustrated with a 3D-printed gyroid seeded with cells. VP-POMaC and low-cost LCD printers make the additive manufacturing of high resolution, elastomeric, and biodegradable constructs widely accessible, paving the way for a myriad of applications in tissue engineering and 3D cell culture as demonstrated here, and possibly in OoC, implants, wearables, and soft robotics.
Collapse
Affiliation(s)
- Vahid Karamzadeh
- Biomedical Engineering DepartmentMcGill UniversityMontrealQCH3A 0G4Canada
- McGill Genome CentreMcGill UniversityMontrealQCH3A 0G4Canada
| | - Molly L. Shen
- Biomedical Engineering DepartmentMcGill UniversityMontrealQCH3A 0G4Canada
- McGill Genome CentreMcGill UniversityMontrealQCH3A 0G4Canada
| | - Hossein Ravanbakhsh
- Biomedical Engineering DepartmentMcGill UniversityMontrealQCH3A 0G4Canada
- McGill Genome CentreMcGill UniversityMontrealQCH3A 0G4Canada
- Department of Biomedical EngineeringThe University of AkronAkronOH44325USA
| | - Ahmad Sohrabi‐Kashani
- Biomedical Engineering DepartmentMcGill UniversityMontrealQCH3A 0G4Canada
- McGill Genome CentreMcGill UniversityMontrealQCH3A 0G4Canada
| | - Sargol Okhovatian
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoONM1C 1A4Canada
| | - Houman Savoji
- Institute of Biomedical EngineeringDepartment of Pharmacology and PhysiologyFaculty of MedicineUniversity of MontrealMontrealQCH3C 3J7Canada
- Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Montreal TransMedTech InstituteMontrealQCH3C 3A7Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoONM1C 1A4Canada
| | - David Juncker
- Biomedical Engineering DepartmentMcGill UniversityMontrealQCH3A 0G4Canada
- McGill Genome CentreMcGill UniversityMontrealQCH3A 0G4Canada
| |
Collapse
|
11
|
Wu Q, Xue R, Zhao Y, Ramsay K, Wang EY, Savoji H, Veres T, Cartmell SH, Radisic M. Automated fabrication of a scalable heart-on-a-chip device by 3D printing of thermoplastic elastomer nanocomposite and hot embossing. Bioact Mater 2024; 33:46-60. [PMID: 38024233 PMCID: PMC10654006 DOI: 10.1016/j.bioactmat.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
The successful translation of organ-on-a-chip devices requires the development of an automated workflow for device fabrication, which is challenged by the need for precise deposition of multiple classes of materials in micro-meter scaled configurations. Many current heart-on-a-chip devices are produced manually, requiring the expertise and dexterity of skilled operators. Here, we devised an automated and scalable fabrication method to engineer a Biowire II multiwell platform to generate human iPSC-derived cardiac tissues. This high-throughput heart-on-a-chip platform incorporated fluorescent nanocomposite microwires as force sensors, produced from quantum dots and thermoplastic elastomer, and 3D printed on top of a polystyrene tissue culture base patterned by hot embossing. An array of built-in carbon electrodes was embedded in a single step into the base, flanking the microwells on both sides. The facile and rapid 3D printing approach efficiently and seamlessly scaled up the Biowire II system from an 8-well chip to a 24-well and a 96-well format, resulting in an increase of platform fabrication efficiency by 17,5000-69,000% per well. The device's compatibility with long-term electrical stimulation in each well facilitated the targeted generation of mature human iPSC-derived cardiac tissues, evident through a positive force-frequency relationship, post-rest potentiation, and well-aligned sarcomeric apparatus. This system's ease of use and its capacity to gauge drug responses in matured cardiac tissue make it a powerful and reliable platform for rapid preclinical drug screening and development.
Collapse
Affiliation(s)
- Qinghua Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Ruikang Xue
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering and The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK
| | - Yimu Zhao
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Kaitlyn Ramsay
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Erika Yan Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Houman Savoji
- Institute of Biomedical Engineering and Department of Pharmacology and Physiology, University of Montreal, Montreal, Quebec, H3T 1J4, Canada
- Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Quebec, H3T 1C5, Canada
- Montreal TransMedTech Institute, Montreal, Quebec, H3T 1J4, Canada
| | - Teodor Veres
- National Research Council of Canada, Boucherville, QC, J4B 6Y4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, M5S 3G8, Canada
| | - Sarah H. Cartmell
- Department of Materials, School of Natural Sciences, Faculty of Science and Engineering and The Henry Royce Institute, Royce Hub Building, The University of Manchester, Manchester, UK
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| |
Collapse
|
12
|
Shahriar SMS, McCarthy AD, Andrabi SM, Su Y, Polavoram NS, John JV, Matis MP, Zhu W, Xie J. Mechanically resilient hybrid aerogels containing fibers of dual-scale sizes and knotty networks for tissue regeneration. Nat Commun 2024; 15:1080. [PMID: 38316777 PMCID: PMC10844217 DOI: 10.1038/s41467-024-45458-x] [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: 09/24/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
The structure and design flexibility of aerogels make them promising for soft tissue engineering, though they tend to come with brittleness and low elasticity. While increasing crosslinking density may improve mechanics, it also imparts brittleness. In soft tissue engineering, resilience against mechanical loads from mobile tissues is paramount. We report a hybrid aerogel that consists of self-reinforcing networks of micro- and nanofibers. Nanofiber segments physically entangle microfiber pillars, allowing efficient stress distribution through the intertwined fiber networks. We show that optimized hybrid aerogels have high specific tensile moduli (~1961.3 MPa cm3 g-1) and fracture energies (~7448.8 J m-2), while exhibiting super-elastic properties with rapid shape recovery (~1.8 s). We demonstrate that these aerogels induce rapid tissue ingrowth, extracellular matrix deposition, and neovascularization after subcutaneous implants in rats. Furthermore, we can apply them for engineering soft tissues via minimally invasive procedures, and hybrid aerogels can extend their versatility to become magnetically responsive or electrically conductive, enabling pressure sensing and actuation.
Collapse
Affiliation(s)
- S M Shatil Shahriar
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Alec D McCarthy
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Syed Muntazir Andrabi
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yajuan Su
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Navatha Shree Polavoram
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Johnson V John
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mitchell P Matis
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale, AZ, 85259, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
13
|
Okhovatian S, Shakeri A, Huyer LD, Radisic M. Elastomeric Polyesters in Cardiovascular Tissue Engineering and Organs-on-a-Chip. Biomacromolecules 2023; 24:4511-4531. [PMID: 37639715 PMCID: PMC10915885 DOI: 10.1021/acs.biomac.3c00387] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cardiovascular tissue constructs provide unique design requirements due to their functional responses to substrate mechanical properties and cyclic stretching behavior of cardiac tissue that requires the use of durable elastic materials. Given the diversity of polyester synthesis approaches, an opportunity exists to develop a new class of biocompatible, elastic, and immunomodulatory cardiovascular polymers. Furthermore, elastomeric polyester materials have the capability to provide tailored biomechanical synergy with native tissue and hence reduce inflammatory response in vivo and better support tissue maturation in vitro. In this review, we highlight underlying chemistry and design strategies of polyester elastomers optimized for cardiac tissue scaffolds. The major advantages of these materials such as their tunable elasticity, desirable biodegradation, and potential for incorporation of bioactive compounds are further expanded. Unique fabrication methods using polyester materials such as micromolding, 3D stamping, electrospinning, laser ablation, and 3D printing are discussed. Moreover, applications of these biomaterials in cardiovascular organ-on-a-chip devices and patches are analyzed. Finally, we outline unaddressed challenges in the field that need further study to enable the impactful translation of soft polyesters to clinical applications.
Collapse
Affiliation(s)
- Sargol Okhovatian
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Locke Davenport Huyer
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
14
|
Wu CA, Zhu Y, Woo YJ. Advances in 3D Bioprinting: Techniques, Applications, and Future Directions for Cardiac Tissue Engineering. Bioengineering (Basel) 2023; 10:842. [PMID: 37508869 PMCID: PMC10376421 DOI: 10.3390/bioengineering10070842] [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: 06/16/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality in the United States. Cardiac tissue engineering is a direction in regenerative medicine that aims to repair various heart defects with the long-term goal of artificially rebuilding a full-scale organ that matches its native structure and function. Three-dimensional (3D) bioprinting offers promising applications through its layer-by-layer biomaterial deposition using different techniques and bio-inks. In this review, we will introduce cardiac tissue engineering, 3D bioprinting processes, bioprinting techniques, bio-ink materials, areas of limitation, and the latest applications of this technology, alongside its future directions for further innovation.
Collapse
Affiliation(s)
- Catherine A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
15
|
Baghersad S, Sathish Kumar A, Kipper MJ, Popat K, Wang Z. Recent Advances in Tissue-Engineered Cardiac Scaffolds-The Progress and Gap in Mimicking Native Myocardium Mechanical Behaviors. J Funct Biomater 2023; 14:jfb14050269. [PMID: 37233379 DOI: 10.3390/jfb14050269] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
Heart failure is the leading cause of death in the US and worldwide. Despite modern therapy, challenges remain to rescue the damaged organ that contains cells with a very low proliferation rate after birth. Developments in tissue engineering and regeneration offer new tools to investigate the pathology of cardiac diseases and develop therapeutic strategies for heart failure patients. Tissue -engineered cardiac scaffolds should be designed to provide structural, biochemical, mechanical, and/or electrical properties similar to native myocardium tissues. This review primarily focuses on the mechanical behaviors of cardiac scaffolds and their significance in cardiac research. Specifically, we summarize the recent development of synthetic (including hydrogel) scaffolds that have achieved various types of mechanical behavior-nonlinear elasticity, anisotropy, and viscoelasticity-all of which are characteristic of the myocardium and heart valves. For each type of mechanical behavior, we review the current fabrication methods to enable the biomimetic mechanical behavior, the advantages and limitations of the existing scaffolds, and how the mechanical environment affects biological responses and/or treatment outcomes for cardiac diseases. Lastly, we discuss the remaining challenges in this field and suggestions for future directions to improve our understanding of mechanical control over cardiac function and inspire better regenerative therapies for myocardial restoration.
Collapse
Affiliation(s)
- Somayeh Baghersad
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Abinaya Sathish Kumar
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Matt J Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Ketul Popat
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
16
|
Liu S, Yu JM, Gan YC, Qiu XZ, Gao ZC, Wang H, Chen SX, Xiong Y, Liu GH, Lin SE, McCarthy A, John JV, Wei DX, Hou HH. Biomimetic natural biomaterials for tissue engineering and regenerative medicine: new biosynthesis methods, recent advances, and emerging applications. Mil Med Res 2023; 10:16. [PMID: 36978167 PMCID: PMC10047482 DOI: 10.1186/s40779-023-00448-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Biomimetic materials have emerged as attractive and competitive alternatives for tissue engineering (TE) and regenerative medicine. In contrast to conventional biomaterials or synthetic materials, biomimetic scaffolds based on natural biomaterial can offer cells a broad spectrum of biochemical and biophysical cues that mimic the in vivo extracellular matrix (ECM). Additionally, such materials have mechanical adaptability, microstructure interconnectivity, and inherent bioactivity, making them ideal for the design of living implants for specific applications in TE and regenerative medicine. This paper provides an overview for recent progress of biomimetic natural biomaterials (BNBMs), including advances in their preparation, functionality, potential applications and future challenges. We highlight recent advances in the fabrication of BNBMs and outline general strategies for functionalizing and tailoring the BNBMs with various biological and physicochemical characteristics of native ECM. Moreover, we offer an overview of recent key advances in the functionalization and applications of versatile BNBMs for TE applications. Finally, we conclude by offering our perspective on open challenges and future developments in this rapidly-evolving field.
Collapse
Affiliation(s)
- Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China
| | - Jiang-Ming Yu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Yan-Chang Gan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China
| | - Xiao-Zhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China
| | - Zhe-Chen Gao
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Huan Wang
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, Guangdong, China.
| | - Shi-Xuan Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325011, Zhejiang, China.
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Si-En Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Alec McCarthy
- Department of Functional Materials, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Johnson V John
- Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68130, USA
| | - Dai-Xu Wei
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China.
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong, 643002, Sichuan, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710127, China.
| | - Hong-Hao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China.
| |
Collapse
|
17
|
Mitchell SM, Pajovich HT, Broas SM, Hugo MM, Banerjee IA. Molecular dynamics simulations and in vitro studies of hybrid decellularized leaf-peptide-polypyrrole composites for potential tissue engineering applications. J Biomol Struct Dyn 2023; 41:1665-1680. [PMID: 34990308 DOI: 10.1080/07391102.2021.2023643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Tissue engineering (TE) aims to repair and regenerate damaged tissue by an assimilation of optimal combination of cells specific to the tissue with an appropriate biomaterial. In this work, a new biomaterial for potential cardiac TE applications was developed by utilizing a combination of in silico studies and in vitro experiments. Molecular dynamics (MD) simulations for the formation of the novel composite prepared from the decellularized leaf components cellulose and pectin along with the VEGF derived peptide (NYLTHRQ) and polypyrrole (PPy) was carried out to assess self-assembly, mechanical properties, and interactions with integrin and NPR-C receptors which are commonly found in cells of cardiac tissue. Results of molecular dynamics simulations indicated the successful formation of stable assemblies. MD simulations also revealed that the scaffold successfully interacted with integrin and NPR-C receptors. As a proof of concept, beet leaves were decellularized (DC) and cross-linked with NYLTHRQ and PPy using layer-by-layer assembly. Decellularization (DC) was confirmed by DNA and protein quantification. Incorporation of the NYLTHRQ peptide and polypyrrole was confirmed by FTIR spectroscopy and SEM imaging. The DC-NYLTHRQ-PPy scaffold was seeded with co-cultured cardiomyocytes and vascular smooth muscle cells. The scaffold promoted cell proliferation and adhesion. Actin and Troponin T immunofluorescence staining showed the presence of these critical cardiomyocyte markers. Thus, for the first time we have developed a decellularized leaf-peptide-PPy composite scaffold by a combination of in silico studies and laboratory analyses that may have potential applications in cardiac TE.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Sarah M Broas
- Department of Chemistry, Fordham University, Bronx, NY, USA
| | - Mindy M Hugo
- Department of Chemistry, Fordham University, Bronx, NY, USA
| | | |
Collapse
|
18
|
Meng Y, Zhai H, Zhou Z, Wang X, Han J, Feng W, Huang Y, Wang Y, Bai Y, Zhou J, Quan D. Three dimensional
printable multi‐arms poly(
CL‐
co
‐TOSUO
) for resilient biodegradable elastomer. JOURNAL OF POLYMER SCIENCE 2023. [DOI: 10.1002/pol.20220700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Affiliation(s)
- Yue Meng
- GD HPPC and PCFM Lab, School of Chemistry Sun Yat‐sen University Guangzhou China
| | - Hong Zhai
- GD HPPC and PCFM Lab, School of Chemistry Sun Yat‐sen University Guangzhou China
| | - Ziting Zhou
- GD Functional Biomaterials Engineering Technology Research Center, Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou China
| | - Xiaoying Wang
- School of Biomedical Engineering Jinan University Guangzhou China
| | - Jiandong Han
- GD Functional Biomaterials Engineering Technology Research Center, Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou China
| | - WenJuan Feng
- GD HPPC and PCFM Lab, School of Chemistry Sun Yat‐sen University Guangzhou China
| | - Yuxin Huang
- GD HPPC and PCFM Lab, School of Chemistry Sun Yat‐sen University Guangzhou China
| | - Yuan Wang
- GD Functional Biomaterials Engineering Technology Research Center, Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou China
| | - Ying Bai
- GD Functional Biomaterials Engineering Technology Research Center, Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou China
| | - Jing Zhou
- GD Functional Biomaterials Engineering Technology Research Center, Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou China
| | - Daping Quan
- GD Functional Biomaterials Engineering Technology Research Center, Guangzhou Key Laboratory of Flexible Electronic Materials and Wearable Devices, School of Materials Science and Engineering Sun Yat‐sen University Guangzhou China
| |
Collapse
|
19
|
Zarei M, Lee G, Lee SG, Cho K. Advances in Biodegradable Electronic Skin: Material Progress and Recent Applications in Sensing, Robotics, and Human-Machine Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2203193. [PMID: 35737931 DOI: 10.1002/adma.202203193] [Citation(s) in RCA: 65] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/13/2022] [Indexed: 06/15/2023]
Abstract
The rapid growth of the electronics industry and proliferation of electronic materials and telecommunications technologies has led to the release of a massive amount of untreated electronic waste (e-waste) into the environment. Consequently, catastrophic environmental damage at the microbiome level and serious human health diseases threaten the natural fate of the planet. Currently, the demand for wearable electronics for applications in personalized medicine, electronic skins (e-skins), and health monitoring is substantial and growing. Therefore, "green" characteristics such as biodegradability, self-healing, and biocompatibility ensure the future application of wearable electronics and e-skins in biomedical engineering and bioanalytical sciences. Leveraging the biodegradability, sustainability, and biocompatibility of natural materials will dramatically influence the fabrication of environmentally friendly e-skins and wearable electronics. Here, the molecular and structural characteristics of biological skins and artificial e-skins are discussed. The focus then turns to the biodegradable materials, including natural and synthetic-polymer-based materials, and their recent applications in the development of biodegradable e-skin in wearable sensors, robotics, and human-machine interfaces (HMIs). Finally, the main challenges and outlook regarding the preparation and application of biodegradable e-skins are critically discussed in a near-future scenario, which is expected to lead to the next generation of biodegradable e-skins.
Collapse
Affiliation(s)
- Mohammad Zarei
- Department of Chemistry, University of Ulsan, Ulsan, 44610, Korea
| | - Giwon Lee
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Korea
| | - Seung Goo Lee
- Department of Chemistry, University of Ulsan, Ulsan, 44610, Korea
| | - Kilwon Cho
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, 37673, Korea
| |
Collapse
|
20
|
Liu C, Campbell SB, Li J, Bannerman D, Pascual-Gil S, Kieda J, Wu Q, Herman PR, Radisic M. High Throughput Omnidirectional Printing of Tubular Microstructures from Elastomeric Polymers. Adv Healthc Mater 2022; 11:e2201346. [PMID: 36165232 PMCID: PMC9742311 DOI: 10.1002/adhm.202201346] [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: 07/04/2022] [Revised: 09/09/2022] [Indexed: 01/28/2023]
Abstract
Bioelastomers are extensively used in biomedical applications due to their desirable mechanical strength, tunable properties, and chemical versatility; however, three-dimensional (3D) printing bioelastomers into microscale structures has proven elusive. Herein, a high throughput omnidirectional printing approach via coaxial extrusion is described that fabricates perfusable elastomeric microtubes of unprecedently small inner diameter (350-550 µm) and wall thickness (40-60 µm). The versatility of this approach is shown through the printing of two different polymeric elastomers, followed by photocrosslinking and removal of the fugitive inner phase. Designed experiments are used to tune the microtube dimensions and stiffness to match that of native ex vivo rat vasculature. This approach affords the fabrication of multiple biomimetic shapes resembling cochlea and kidney glomerulus and affords facile, high-throughput generation of perfusable structures that can be seeded with endothelial cells for biomedical applications. Post-printing laser micromachining is performed to generate micro-sized holes (520 µm) in the tube wall to tune microstructure permeability. Importantly, for organ-on-a-chip applications, the described approach takes only 3.6 min to print microtubes (without microholes) over an entire 96-well plate device, in contrast to comparable hole-free structures that take between 1.5 and 6.5 days to fabricate using a manual 3D stamping approach.
Collapse
Affiliation(s)
- Chuan Liu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Scott B. Campbell
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jianzhao Li
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Dawn Bannerman
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Simon Pascual-Gil
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Qinghua Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Peter R. Herman
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Nahak BK, Mishra A, Preetam S, Tiwari A. Advances in Organ-on-a-Chip Materials and Devices. ACS APPLIED BIO MATERIALS 2022; 5:3576-3607. [PMID: 35839513 DOI: 10.1021/acsabm.2c00041] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The organ-on-a-chip (OoC) paves a way for biomedical applications ranging from preclinical to clinical translational precision. The current trends in the in vitro modeling is to reduce the complexity of human organ anatomy to the fundamental cellular microanatomy as an alternative of recreating the entire cell milieu that allows systematic analysis of medicinal absorption of compounds, metabolism, and mechanistic investigation. The OoC devices accurately represent human physiology in vitro; however, it is vital to choose the correct chip materials. The potential chip materials include inorganic, elastomeric, thermoplastic, natural, and hybrid materials. Despite the fact that polydimethylsiloxane is the most commonly utilized polymer for OoC and microphysiological systems, substitute materials have been continuously developed for its advanced applications. The evaluation of human physiological status can help to demonstrate using noninvasive OoC materials in real-time procedures. Therefore, this Review examines the materials used for fabricating OoC devices, the application-oriented pros and cons, possessions for device fabrication and biocompatibility, as well as their potential for downstream biochemical surface alteration and commercialization. The convergence of emerging approaches, such as advanced materials, artificial intelligence, machine learning, three-dimensional (3D) bioprinting, and genomics, have the potential to perform OoC technology at next generation. Thus, OoC technologies provide easy and precise methodologies in cost-effective clinical monitoring and treatment using standardized protocols, at even personalized levels. Because of the inherent utilization of the integrated materials, employing the OoC with biomedical approaches will be a promising methodology in the healthcare industry.
Collapse
Affiliation(s)
- Bishal Kumar Nahak
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Anshuman Mishra
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| |
Collapse
|
22
|
Polonchuk L, Gentile C. Current state and future of 3D bioprinted models for cardiovascular research and drug development. ADMET AND DMPK 2022; 9:231-242. [PMID: 35300373 PMCID: PMC8920100 DOI: 10.5599/admet.951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
In the last decade, 3D bioprinting technology has emerged as an innovative tissue engineering approach for regenerative medicine and drug development. This article aims at providing an overview about the most commonly used bioengineered tissues, focusing on 3D bioprinted cardiac cells and how they have been utilized for drug discovery and development. The review describes that, while this field is still developing, cardiovascular research may benefit from laboratory-engineered heart tissues built of specific cell types with precise 3D architecture mimicking the native cardiac microenvironment. It also describes the role played by regulatory agencies and potential commercialization pathways for direct translation from the bench to the bedside of studies using 3D bioprinted cardiac tissues.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Carmine Gentile
- Sydney Medical School, The University of Sydney, Australia.,School of Biomedical Engineering, University of Technology Sydney, Australia
| |
Collapse
|
23
|
Gao Y, Xue J, Zhang L, Wang Z. Synthesis of bio-based polyester elastomers and evaluation of the in vivo biocompatibility and biodegradability as biomedical materials. Biomater Sci 2022; 10:3924-3934. [DOI: 10.1039/d2bm00436d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biodegradable polyester elastomers have found wide applications in the tissue engineering field. In this study, all bio-based polyester elastomer (BPE) is synthesized from five bio-based monomers; and the in vivo...
Collapse
|
24
|
Wang EY, Smith J, Radisic M. Design and Fabrication of Biological Wires for Cardiac Fibrosis Disease Modeling. Methods Mol Biol 2022; 2485:175-190. [PMID: 35618906 DOI: 10.1007/978-1-0716-2261-2_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Extensive progress has been made in developing engineered models for elucidating human cardiac disease. Cardiac fibrosis is often associated with all forms of cardiac disease and has a direct deleterious effect on cardiac function. As currently there is no effective therapeutic strategy specifically designed to target fibrosis, in vitro diagnostic platforms for drug testing have generated significant interest. In this context, we have developed an innovative approach to generate human cardiac fibrotic tissues on Biowire II platform and established a compound screening system. The disease model is constructed to recapitulate contractile, biomechanical, and electrophysiological complexities of fibrotic myocardium. Additionally, an integrated model with fibrotic and healthy cardiac tissues coupled together can be created to mimic focal fibrosis. The methods for constructing the Biowire fibrotic model will be described here.
Collapse
Affiliation(s)
- Erika Yan Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Jacob Smith
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
25
|
Turner B, Ramesh S, Menegatti S, Daniele M. Resorbable elastomers for implantable medical devices: highlights and applications. POLYM INT 2021. [DOI: 10.1002/pi.6349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Brendan Turner
- Joint Department of Biomedical Engineering North Carolina State University and University of Chapel Hill Raleigh NC USA
- Department of Chemical and Biomolecular Engineering North Carolina State University Raleigh NC USA
| | - Srivatsan Ramesh
- Department of Chemical and Biomolecular Engineering North Carolina State University Raleigh NC USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering North Carolina State University Raleigh NC USA
| | - Michael Daniele
- Joint Department of Biomedical Engineering North Carolina State University and University of Chapel Hill Raleigh NC USA
- Department of Electrical and Computer Engineering North Carolina State University Raleigh NC USA
| |
Collapse
|
26
|
Li X, Qi X, Cai YM, Sun Y, Wen R, Zhang R, Johansson J, Meng Q, Chen G. Customized Flagelliform Spidroins Form Spider Silk-like Fibers at pH 8.0 with Outstanding Tensile Strength. ACS Biomater Sci Eng 2021; 8:119-127. [PMID: 34908395 PMCID: PMC8753598 DOI: 10.1021/acsbiomaterials.1c01354] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Spider flagelliform silk shows the best extensibility among various types of silk, but its biomimetic preparation has not been much studied. Herein, five customized flagelliform spidroins (FlSps: S and NTDFl-Sn-CTDFl, n = 1-4), in which the repetitive region (S) and N-/C- terminal domains (NTDFl and CTDFl) are from the same spidroin and spider species, were produced recombinantly. The recombinant spidroins with terminal domains were able to form silk-like fibers with diameters of ∼5 μm by manual pulling at pH 8.0, where the secondary structure transformation occurred. The silk-like fibers from NTDFl-S4-CTDFl showed the highest tensile strength (∼250 MPa), while those ones with 1-3 S broke at a similar stress (∼180 MPa), suggesting that increasing the amounts of the repetitive region can improve the tensile strength, but a certain threshold might need to be reached. This study shows successful preparation of flagelliform silk-like fibers with good mechanical properties, providing general insights into efficient biomimetic preparations of spider silks.
Collapse
Affiliation(s)
- Xue Li
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University Cancer Center, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, 200092 Shanghai, China.,Institute of Biological Sciences and Biotechnology, Donghua University, 201620 Shanghai, China
| | - Xingmei Qi
- The Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yu-Ming Cai
- Institute for Life Sciences, University of Southampton, SO17 1BJ Southampton, Hampshire, U.K
| | - Yuan Sun
- Institute of Biological Sciences and Biotechnology, Donghua University, 201620 Shanghai, China
| | - Rui Wen
- Institute of Biological Sciences and Biotechnology, Donghua University, 201620 Shanghai, China
| | - Rui Zhang
- Department of Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Jan Johansson
- Department of Biosciences and Nutrition, Karolinska Institutet, 14157 Huddinge, Sweden
| | - Qing Meng
- Institute of Biological Sciences and Biotechnology, Donghua University, 201620 Shanghai, China
| | - Gefei Chen
- Department of Biosciences and Nutrition, Karolinska Institutet, 14157 Huddinge, Sweden
| |
Collapse
|
27
|
Zhang P, Shao N, Qin L. Recent Advances in Microfluidic Platforms for Programming Cell-Based Living Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005944. [PMID: 34270839 DOI: 10.1002/adma.202005944] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/20/2020] [Indexed: 06/13/2023]
Abstract
Cell-based living materials, including single cells, cell-laden fibers, cell sheets, organoids, and organs, have attracted intensive interests owing to their widespread applications in cancer therapy, regenerative medicine, drug development, and so on. Significant progress in materials, microfabrication, and cell biology have promoted the development of numerous promising microfluidic platforms for programming these cell-based living materials with a high-throughput, scalable, and efficient manner. In this review, the recent progress of novel microfluidic platforms for programming cell-based living materials is presented. First, the unique features, categories, and materials and related fabrication methods of microfluidic platforms are briefly introduced. From the viewpoint of the design principles of the microfluidic platforms, the recent significant advances of programming single cells, cell-laden fibers, cell sheets, organoids, and organs in turns are then highlighted. Last, by providing personal perspectives on challenges and future trends, this review aims to motivate researchers from the fields of materials and engineering to work together with biologists and physicians to promote the development of cell-based living materials for human healthcare-related applications.
Collapse
Affiliation(s)
- Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Ning Shao
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, 10065, USA
| |
Collapse
|
28
|
Lu RXZ, Radisic M. Organ-on-a-chip platforms for evaluation of environmental nanoparticle toxicity. Bioact Mater 2021; 6:2801-2819. [PMID: 33665510 PMCID: PMC7900603 DOI: 10.1016/j.bioactmat.2021.01.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Despite showing a great promise in the field of nanomedicine, nanoparticles have gained a significant attention from regulatory agencies regarding their possible adverse health effects upon environmental exposure. Whether those nanoparticles are generated through intentional or unintentional means, the constant exposure to nanomaterials can inevitably lead to unintended consequences based on epidemiological data, yet the current understanding of nanotoxicity is insufficient relative to the rate of their emission in the environment and the lack of predictive platforms that mimic the human physiology. This calls for a development of more physiologically relevant models, which permit the comprehensive and systematic examination of toxic properties of nanoparticles. With the advancement in microfabrication techniques, scientists have shifted their focus on the development of an engineered system that acts as an intermediate between a well-plate system and animal models, known as organ-on-a-chips. The ability of organ-on-a-chip models to recapitulate in vivo like microenvironment and responses offers a new avenue for nanotoxicological research. In this review, we aim to provide overview of assessing potential risks of nanoparticle exposure using organ-on-a-chip systems and their potential to delineate biological mechanisms of epidemiological findings.
Collapse
Affiliation(s)
- Rick Xing Ze Lu
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- The Heart and Stroke/Richard Lewar Centre of Excellence, Toronto, ON, Canada
| |
Collapse
|
29
|
Wang Z, He X, He T, Zhao J, Wang S, Peng S, Yang D, Ye L. Polymer Network Editing of Elastomers for Robust Underwater Adhesion and Tough Bonding to Diverse Surfaces. ACS APPLIED MATERIALS & INTERFACES 2021; 13:36527-36537. [PMID: 34313126 DOI: 10.1021/acsami.1c09239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tough adhesives with robust adhesion are desperately needed for biomedical and technological applications. However, it is extremely challenging to engineer tough and durable adhesives that are simple to make yet also exhibit strong underwater adhesion as well as tough bonding to diverse surfaces. Here, we report bioinspired elastomers based on water-immiscible polydiolcitrates, where their tough mechanical properties, robust underwater adhesion (80 kPa), and tough bonding performance (with an interfacial toughness >1000 J m-2 and a shear and tensile strength >0.5 MPa) to diverse solid materials (glass, ceramics, and steel) are actuated by the incorporation of trace amounts of additives. The additives could edit the polymer networks during the elastomer polymerization by dramatically regulating the cross-linking structures of covalent and reversible bonds, the length of polymer chains, and the hydrophobic and hydrophilic motifs, which markedly tuned the mechanical and adhesive properties of the bioelastomers. We also demonstrate versatile applications of the durable elastomers, as tough flexible joints for solid materials, superglue, tissue sealants, hemostatic dressing, and wound repair.
Collapse
Affiliation(s)
- Zhenming Wang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, 3th Section, South Renmin Road, Wuhou District, Chengdu 610041, China
| | - Xiaoqin He
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Tongzhong He
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Jin Zhao
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Shang Wang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Songlin Peng
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Dazhi Yang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, 3th Section, South Renmin Road, Wuhou District, Chengdu 610041, China
| |
Collapse
|
30
|
Rafatian N, Vizely K, Al Asafen H, Korolj A, Radisic M. Drawing Inspiration from Developmental Biology for Cardiac Tissue Engineers. Adv Biol (Weinh) 2021; 5:e2000190. [PMID: 34008910 DOI: 10.1002/adbi.202000190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/21/2020] [Indexed: 12/17/2022]
Abstract
A sound understanding of developmental biology is part of the foundation of effective stem cell-derived tissue engineering. Here, the key concepts of cardiac development that are successfully applied in a bioinspired approach to growing engineered cardiac tissues, are reviewed. The native cardiac milieu is studied extensively from embryonic to adult phenotypes, as it provides a resource of factors, mechanisms, and protocols to consider when working toward establishing living tissues in vitro. It begins with the various cell types that constitute the cardiac tissue. It is discussed how myocytes interact with other cell types and their microenvironment and how they change over time from the embryonic to the adult states, with a view on how such changes affect the tissue function and may be used in engineered tissue models. Key embryonic signaling pathways that have been leveraged in the design of culture media and differentiation protocols are presented. The cellular microenvironment, from extracellular matrix chemical and physical properties, to the dynamic mechanical and electrical forces that are exerted on tissues is explored. It is shown that how such microenvironmental factors can inform the design of biomaterials, scaffolds, stimulation bioreactors, and maturation readouts, and suggest considerations for ongoing biomimetic advancement of engineered cardiac tissues and regeneration strategies for the future.
Collapse
Affiliation(s)
- Naimeh Rafatian
- Toronto General Research Institute, Toronto, Ontario, M5G 2C4, Canada
| | - Katrina Vizely
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Hadel Al Asafen
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Milica Radisic
- Toronto General Research Institute, Toronto, Ontario, M5G 2C4, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomaterials Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
31
|
Responsive Polyesters with Alkene and Carboxylic Acid Side-Groups for Tissue Engineering Applications. Polymers (Basel) 2021; 13:polym13101636. [PMID: 34070123 PMCID: PMC8158382 DOI: 10.3390/polym13101636] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 11/17/2022] Open
Abstract
Main chain polyesters have been extensively used in the biomedical field. Despite their many advantages, including biocompatibility, biodegradability, and others, these materials are rather inert and lack specific functionalities which will endow them with additional biological and responsive properties. In this work, novel pH-responsive main chain polyesters have been prepared by a conventional condensation polymerization of a vinyl functionalized diol with a diacid chloride, followed by a photo-induced thiol-ene click reaction to attach functional carboxylic acid side-groups along the polymer chains. Two different mercaptocarboxylic acids were employed, allowing to vary the alkyl chain length of the polymer pendant groups. Moreover, the degree of modification, and as a result, the carboxylic acid content of the polymers, was easily tuned by varying the irradiation time during the click reaction. Both these parameters, were shown to strongly influence the responsive behavior of the polyesters, which presented adjustable pKα values and water solubilities. Finally, the difunctional polyesters bearing the alkene and carboxylic acid functionalities enabled the preparation of cross-linked polyester films by chemically linking the pendant vinyl bonds on the polymer side groups. The biocompatibility of the cross-linked polymers films was assessed in L929 fibroblast cultures and showed that the cell viability, proliferation, and attachment were greatly promoted on the polyester surface, bearing the shorter alkyl chain length side groups and the higher fraction of carboxylic acid functionalities.
Collapse
|
32
|
Kirillova A, Yeazel TR, Asheghali D, Petersen SR, Dort S, Gall K, Becker ML. Fabrication of Biomedical Scaffolds Using Biodegradable Polymers. Chem Rev 2021; 121:11238-11304. [PMID: 33856196 DOI: 10.1021/acs.chemrev.0c01200] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Degradable polymers are used widely in tissue engineering and regenerative medicine. Maturing capabilities in additive manufacturing coupled with advances in orthogonal chemical functionalization methodologies have enabled a rapid evolution of defect-specific form factors and strategies for designing and creating bioactive scaffolds. However, these defect-specific scaffolds, especially when utilizing degradable polymers as the base material, present processing challenges that are distinct and unique from other classes of materials. The goal of this review is to provide a guide for the fabrication of biodegradable polymer-based scaffolds that includes the complete pathway starting from selecting materials, choosing the correct fabrication method, and considering the requirements for tissue specific applications of the scaffold.
Collapse
Affiliation(s)
- Alina Kirillova
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Taylor R Yeazel
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Darya Asheghali
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Shannon R Petersen
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Sophia Dort
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Ken Gall
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Matthew L Becker
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States.,Department of Chemistry, Duke University, Durham, North Carolina 27708, United States.,Departments of Biomedical Engineering and Orthopaedic Surgery, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
33
|
A well plate-based multiplexed platform for incorporation of organoids into an organ-on-a-chip system with a perfusable vasculature. Nat Protoc 2021; 16:2158-2189. [PMID: 33790475 DOI: 10.1038/s41596-020-00490-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023]
Abstract
Owing to their high spatiotemporal precision and adaptability to different host cells, organ-on-a-chip systems are showing great promise in drug discovery, developmental biology studies and disease modeling. However, many current micro-engineered biomimetic systems are limited in technological application because of culture media mixing that does not allow direct incorporation of techniques from stem cell biology, such as organoids. Here, we describe a detailed alternative method to cultivate millimeter-scale functional vascularized tissues on a biofabricated platform, termed 'integrated vasculature for assessing dynamic events', that enables facile incorporation of organoid technology. Utilizing the 3D stamping technique with a synthetic polymeric elastomer, a scaffold termed 'AngioTube' is generated with a central microchannel that has the mechanical stability to support a perfusable vascular system and the self-assembly of various parenchymal tissues. We demonstrate an increase in user familiarity and content analysis by situating the scaffold on a footprint of a 96-well plate. Uniquely, the platform can be used for facile connection of two or more tissue compartments in series through a common vasculature. Built-in micropores enable the studies of cell invasion involved in both angiogenesis and metastasis. We describe how this protocol can be applied to create both vascularized cardiac and hepatic tissues, metastatic breast cancer tissue and personalized pancreatic cancer tissue through incorporation of patient-derived organoids. Platform assembly to populating the scaffold with cells of interest into perfusable functional vascularized tissue will require 12-14 d and an additional 4 d if pre-polymer and master molds are needed.
Collapse
|
34
|
Wang Z, Zhao J, Tang W, He T, Wang S, He X, Chen Y, Yang D, Peng S. Robust Underwater Adhesives Based on Dynamic Hydrophilic and Hydrophobic Moieties to Diverse Surfaces. ACS APPLIED MATERIALS & INTERFACES 2021; 13:3435-3444. [PMID: 33405512 DOI: 10.1021/acsami.0c20186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Underwater adhesives (UAs) have promising applications in diverse areas. However, traditional UAs have several drawbacks such as weak and irreversible adhesion behaviors as well as poor performance in biological environments. To address these challenges, we engineered a novel synthetic adhesive based on dynamic hydrophilic and hydrophobic moieties, which shows very strong underwater adhesion strength (30-110 kPa) and debonding energy (20-100 J/m2) to diverse substrates. Interestingly, the UAs could also be switched reversibly and repeatedly by the dynamic exchange of hydrophilic and hydrophobic moieties under alternating temperatures. We also demonstrate the versatile functions and practical value of the UAs for clinical applications as tissue sealants and hemostatic dressing in emergency rescue operations. This general and efficient strategy may be generalized to develop additional next generation UAs for many emerging technological and medical applications.
Collapse
Affiliation(s)
- Zhenming Wang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jin Zhao
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Wanze Tang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Tongzhong He
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Shang Wang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Xiaoqin He
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Yang Chen
- Department of Orthopaedics, The First People's Hospital of Foshan, Foshan, Guangdong 528000, China
| | - Dazhi Yang
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Songlin Peng
- Department of Spine Surgery and Institute for Orthopaedic Research, Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University Second College of Medicine, Shenzhen 518020, China
| |
Collapse
|
35
|
Santos Rosalem G, Gonzáles Torres LA, de Las Casas EB, Mathias FAS, Ruiz JC, Carvalho MGR. Microfluidics and organ-on-a-chip technologies: A systematic review of the methods used to mimic bone marrow. PLoS One 2020; 15:e0243840. [PMID: 33306749 PMCID: PMC7732112 DOI: 10.1371/journal.pone.0243840] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/29/2020] [Indexed: 12/15/2022] Open
Abstract
Bone marrow (BM) is an organ responsible for crucial processes in living organs, e. g., hematopoiesis. In recent years, Organ-on-a-Chip (OoC) devices have been used to satisfy the need for in vitro systems that better mimic the phenomena occurring in the BM microenvironment. Given the growing interest in these systems and the diversity of developed devices, an integrative systematic literature review is required. We have performed this review, following the PRISMA method aiming to identify the main characteristics and assess the effectiveness of the devices that were developed to represent the BM. A search was performed in the Scopus, PubMed, Web of Science and Science Direct databases using the keywords (("bone marrow" OR "hematopoietic stem cells" OR "haematopoietic stem cells") AND ("organ in a" OR "lab on a chip" OR "microfluidic" OR "microfluidic*" OR ("bioreactor" AND "microfluidic*"))). Original research articles published between 2009 and 2020 were included in the review, giving a total of 21 papers. The analysis of these papers showed that their main purpose was to study BM cells biology, mimic BM niches, model pathological BM, and run drug assays. Regarding the fabrication protocols, we have observed that polydimethylsiloxane (PDMS) material and soft lithography method were the most commonly used. To reproduce the microenvironment of BM, most devices used the type I collagen and alginate. Peristaltic and syringe pumps were mostly used for device perfusion. Regarding the advantages compared to conventional methods, there were identified three groups of OoC devices: perfused 3D BM; co-cultured 3D BM; and perfused co-cultured 3D BM. Cellular behavior and mimicking their processes and responses were the mostly commonly studied parameters. The results have demonstrated the effectiveness of OoC devices for research purposes compared to conventional cell cultures. Furthermore, the devices have a wide range of applicability and the potential to be explored.
Collapse
Affiliation(s)
- Gabriel Santos Rosalem
- Mechanical Engineering Graduate Program, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | - Jeronimo Conceição Ruiz
- Biosystems and Genomics Group, René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
- Graduate Program in Computational and Systems Biology of the Institute Oswaldo Cruz (PGBCS/IOC/Fiocruz), Rio de Janeiro, Brazil
| | | |
Collapse
|
36
|
Lai Benjamin FL, Lu Rick X, Hu Y, Davenport HL, Dou W, Wang EY, Radulovich N, Tsao MS, Sun Y, Radisic M. Recapitulating pancreatic tumor microenvironment through synergistic use of patient organoids and organ-on-a-chip vasculature. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2000545. [PMID: 33692660 PMCID: PMC7939064 DOI: 10.1002/adfm.202000545] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Tumor progression relies heavily on the interaction between the neoplastic epithelial cells and their surrounding stromal partners. This cell cross-talk affects stromal development, and ultimately the heterogeneity impacts drug efflux and efficacy. To mimic this evolving paradigm, we have micro-engineered a three-dimensional (3D) vascularized pancreatic adenocarcinoma tissue in a tri-culture system composed of patient derived pancreatic organoids, primary human fibroblasts and endothelial cells on a perfusable InVADE platform situated in a 96-well plate. Uniquely, through synergistic engineering we combined the benefits of cellular fidelity of patient tumor derived organoids with the addressability of a plastic organ-on-a-chip platform. Validation of this platform included demonstrating the growth of pancreatic tumor organoids by monitoring the change in metabolic activity of the tissue. Investigation of tumor microenvironmental behavior highlighted the role of fibroblasts in symbiosis with patient organoid cells, resulting in a six-fold increase of collagen deposition and a corresponding increase in tissue stiffness in comparison to fibroblast free controls. The value of a perfusable vascular network was evident in drug screening, as perfusion of gemcitabine into a stiffened matrix did not show the dose-dependent effects on tumor viability as those under static conditions. These findings demonstrate the importance of studying the dynamic synergistic relationship between patient cells with stromal fibroblasts, in a 3D perfused vascular network, to accurately understand and recapitulate the tumor microenvironment.
Collapse
Affiliation(s)
- F L Lai Benjamin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - X Lu Rick
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Yangshuo Hu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Huyer Locke Davenport
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Wenkun Dou
- Material Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Erika Y Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Nikolina Radulovich
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ming S Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yu Sun
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Material Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Bannerman AD, Davenport Huyer L, Montgomery M, Zhao N, Velikonja C, Bender TP, Radisic M. Elastic Biomaterial Scaffold with Spatially Varying Adhesive Design. ADVANCED BIOSYSTEMS 2020; 4:e2000046. [PMID: 32567253 PMCID: PMC7665997 DOI: 10.1002/adbi.202000046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/08/2020] [Indexed: 12/20/2022]
Abstract
In order to secure biomaterials to tissue surfaces, sutures or glues are commonly used. Of interest is the development of a biomaterial patch for applications in tissue engineering and regeneration that incorporates an adhesive component to simplify patch application and ensure sufficient adhesion. A separate region dedicated to fulfilling the specific requirements of an application such as mechanical support or tissue delivery is also desirable. Here, the design and fabrication of a unique patch are presented with distinct regions for adhesion and function, resulting in a biomaterial patch resembling the Band-Aid. The adhesive region contains a novel polymer, synthesized to incorporate a molecule capable of adhesion to tissue, dopamine. The desired polymer composition for patch development is selected based on chemical assessment and evaluation of key physical properties such as swelling and elastic modulus, which are tailored for use in soft tissue applications. The selected polymer formulation, referred to as the adhesive patch (AP) polymer, demonstrates negligible cytotoxicity and improves adhesive capability to rat cardiac tissue compared to currently used patch materials. Finally, the AP polymer is used in the patch, designed to possess distinct adhesive and nonadhesive domains, presenting a novel design for the next generation of biomaterials.
Collapse
Affiliation(s)
- A Dawn Bannerman
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Locke Davenport Huyer
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Miles Montgomery
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Nicholas Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Claire Velikonja
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Timothy P Bender
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| |
Collapse
|
38
|
Ding C, Chen X, Kang Q, Yan X. Biomedical Application of Functional Materials in Organ-on-a-Chip. Front Bioeng Biotechnol 2020; 8:823. [PMID: 32793573 PMCID: PMC7387427 DOI: 10.3389/fbioe.2020.00823] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/29/2020] [Indexed: 01/06/2023] Open
Abstract
The organ-on-a-chip (OOC) technology has been utilized in a lot of biomedical fields such as fundamental physiological and pharmacological researches. Various materials have been introduced in OOC and can be broadly classified into inorganic, organic, and hybrid materials. Although PDMS continues to be the preferred material for laboratory research, materials for OOC are constantly evolving and progressing, and have promoted the development of OOC. This mini review provides a summary of the various type of materials for OOC systems, focusing on the progress of materials and related fabrication technologies within the last 5 years. The advantages and drawbacks of these materials in particular applications are discussed. In addition, future perspectives and challenges are also discussed.
Collapse
Affiliation(s)
- Chizhu Ding
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Xiang Chen
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Qinshu Kang
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Xianghua Yan
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| |
Collapse
|
39
|
Firoozi N, Kang Y. Immobilization of FGF on Poly(xylitol dodecanedioic Acid) Polymer for Tissue Regeneration. Sci Rep 2020; 10:10419. [PMID: 32591607 PMCID: PMC7320172 DOI: 10.1038/s41598-020-67261-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) plays a vital role in the repair and regeneration of most tissues. However, its low stability, short half-life, and rapid inactivation by enzymes in physiological conditions affect their clinical applications. Therefore, to increase the effectiveness of growth factors and to improve tissue regeneration, we developed an elastic polymeric material poly(xylitol dodecanedioic acid) (PXDDA) and loaded FGF on the PXDDA for sustained drug delivery. In this study, we used a simple dopamine coating method to load FGF on the surface of PXDDA polymeric films. The polydopamine-coated FGF-loaded PXDDA samples were then characterized using FTIR and XRD. The in vitro drug release profile of FGF from PXDDA film and cell growth behavior were measured. Results showed that the polydopamine layer coated on the surface of the PXDDA film enhanced the immobilization of FGF and controlled its sustained release. Human fibroblast cells attachment and proliferation on FGF-immobilized PXDDA films were much higher than the other groups without coatings or FGF loading. Based on our results, the surface modification procedure with immobilizing growth factors shows excellent application potential in tissue regeneration.
Collapse
Affiliation(s)
- Negar Firoozi
- Department of Ocean & Mechanical Engineering, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida, 33431, United States
| | - Yunqing Kang
- Department of Ocean & Mechanical Engineering, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida, 33431, United States.
- Department of Biomedical Science, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida, 33431, United States.
- Integrative Biology Ph.D. Program, Department of Biological Science, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida, 33431, United States.
| |
Collapse
|
40
|
Gradinaru LM, Barbalata-Mandru M, Drobota M, Aflori M, Spiridon M, Gradisteanu Pircalabioru G, Bleotu C, Butnaru M, Vlad S. Preparation and Evaluation of Nanofibrous Hydroxypropyl Cellulose and β-Cyclodextrin Polyurethane Composite Mats. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E754. [PMID: 32326486 PMCID: PMC7221721 DOI: 10.3390/nano10040754] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/04/2020] [Accepted: 04/13/2020] [Indexed: 02/02/2023]
Abstract
A series of nanofibrous composite mats based on polyurethane urea siloxane (PUUS), hydroxypropyl cellulose (HPC) and β-cyclodextrin (β-CD) was prepared using electrospinning technique. PUUS was synthesized by two steps solution polymerization procedure from polytetramethylene ether glycol (PTMEG), dimethylol propionic acid (DMPA), 4,4'-diphenylmethane diisocyanate (MDI) and 1,3-bis-(3-aminopropyl) tetramethyldisiloxane (BATD) as chain extender. Then, the composites were prepared by blending PUUS with HPC or βCD in a ratio of 9:1 (w/w), in 15% dimethylformamide (DMF). The PUUS and PUUS based composite solutions were used for preparation of nanofibrous mats. In order to identify the potential applications, different techniques were used to evaluate the chemical structure (Fourier transform infrared-attenuated total reflectance spectroscopy-FTIR-ATR), morphological structure (Scanning electron microscopy-SEM and Atomic force microscopy-AFM), surface properties (contact angle, dynamic vapors sorption-DVS), mechanical characteristics (tensile tests), thermal (differential scanning calorimetry-DSC) and some preliminary tests for biocompatibility and microbial adhesion.
Collapse
Affiliation(s)
- Luiza Madalina Gradinaru
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
| | - Mihaela Barbalata-Mandru
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
| | - Mioara Drobota
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
| | - Magdalena Aflori
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
| | - Maria Spiridon
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
| | | | - Coralia Bleotu
- Sanimed International Impex S.R.L, 70F Bucuresti—Măgurele, 051434 Bucuresti, Romania; (G.G.P.); (C.B.)
- “Stefan S Nicolau” Institute of Virology, 285 Mihai Bravu, 030304 Bucuresti, Romania
| | - Maria Butnaru
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
- Faculty of Medical Bioengineering, “Gr. T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Stelian Vlad
- “P. Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania; (M.B.-M.); (M.D.); (M.A.); (M.S.); (M.B.)
| |
Collapse
|
41
|
De Santis M, Cacciotti I. Wireless implantable and biodegradable sensors for postsurgery monitoring: current status and future perspectives. NANOTECHNOLOGY 2020; 31:252001. [PMID: 32101794 DOI: 10.1088/1361-6528/ab7a2d] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In in vivo postsurgery monitoring, the use of wireless biodegradable implantable sensors has gained and is gaining a lot of interest, particularly in cases of monitoring for a short period of time. The employment of biodegradable materials allows the circumvention of secondary surgery for device removal. Additionally, the use of wireless communication for data elaboration avoids the need for transcutaneous wires. As such, it is possible to prevent possible inflammation and infections associated with long-term implants which are not wireless. It is expected that microfabricated biodegradable sensors will have a strong impact in acute or transient biomedical applications. However, the design of such high-performing electronic systems, both fully biodegradable and wireless, is very complex, particularly at small scales. The associated technologies are still in their infancy and should be more deeply and extensively investigated in animal models and, successively, in humans, before being clinically implemented. In this context, the present review aims to provide a complete overview of wireless biodegradable implantable sensors, covering the vital signs to be monitored, the wireless technologies involved, and the biodegradable materials used for the production of the devices, as well as designed devices and their applications. In particular, both their advantages and drawbacks are highlighted, and the key challenges faced, mainly associated with fabrication techniques, and control over degradation kinetics and biocompatibility of the device, are reported and discussed.
Collapse
Affiliation(s)
- Michele De Santis
- University of Rome 'Niccolò Cusano', Engineering Department, Via Don Carlo Gnocchi 3, 00166 Rome, Italy
| | | |
Collapse
|
42
|
Savoji H, Davenport Huyer L, Mohammadi MH, Lun Lai BF, Rafatian N, Bannerman D, Shoaib M, Bobicki ER, Ramachandran A, Radisic M. 3D Printing of Vascular Tubes Using Bioelastomer Prepolymers by Freeform Reversible Embedding. ACS Biomater Sci Eng 2020; 6:1333-1343. [PMID: 33455372 DOI: 10.1021/acsbiomaterials.9b00676] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bioelastomers have been extensively used in tissue engineering applications because of favorable mechanical stability, tunable properties, and chemical versatility. As these materials generally possess low elastic modulus and relatively long gelation time, it is challenging to 3D print them using traditional techniques. Instead, the field of 3D printing has focused preferentially on hydrogels and rigid polyester materials. To develop a versatile approach for 3D printing of elastomers, we used freeform reversible embedding of suspended prepolymers. A family of novel fast photocrosslinakble bioelastomer prepolymers were synthesized from dimethyl itaconate, 1,8-octanediol, and triethyl citrate. Tensile testing confirmed their elastic properties with Young's moduli in the range of 11-53 kPa. These materials supported cultivation of viable cells and enabled adhesion and proliferation of human umbilical vein endothelial cells. Tubular structures were created by embedding the 3D printed microtubes within a secondary hydrogel that served as a temporary support. Upon photocrosslinking and porogen leaching, the polymers were permeable to small molecules (TRITC-dextran). The polymer microtubes were assembled on the 96-well plates custom made by hot-embossing, as a tool to connect multiple organs-on-a-chip. The endothelialization of the tubes was performed to confirm that these microtubes can be utilized as vascular tubes to support parenchymal tissues seeded on them.
Collapse
Affiliation(s)
- Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College Street, Toronto, Ontario M5S 3G9, Canada.,Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Locke Davenport Huyer
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College Street, Toronto, Ontario M5S 3G9, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Mohammad Hossein Mohammadi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Benjamin Fook Lun Lai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College Street, Toronto, Ontario M5S 3G9, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Mohammad Shoaib
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Erin R Bobicki
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Arun Ramachandran
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College Street, Toronto, Ontario M5S 3G9, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada.,Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
43
|
Biodegradable polymers: a cure for the planet, but a long way to go. JOURNAL OF POLYMER RESEARCH 2020. [DOI: 10.1007/s10965-020-2004-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Zaokari Y, Persaud A, Ibrahim A. Biomaterials for Adhesion in Orthopedic Applications: A Review. ENGINEERED REGENERATION 2020. [DOI: 10.1016/j.engreg.2020.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
45
|
Davenport Huyer L, Bannerman AD, Wang Y, Savoji H, Knee‐Walden EJ, Brissenden A, Yee B, Shoaib M, Bobicki E, Amsden BG, Radisic M. One-Pot Synthesis of Unsaturated Polyester Bioelastomer with Controllable Material Curing for Microscale Designs. Adv Healthc Mater 2019; 8:e1900245. [PMID: 31313890 DOI: 10.1002/adhm.201900245] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/13/2019] [Indexed: 12/17/2022]
Abstract
Synthetic polyester elastomeric constructs have become increasingly important for a range of healthcare applications, due to tunable soft elastic properties that mimic those of human tissues. A number of these constructs require intricate mechanical design to achieve a tunable material with controllable curing. Here, the synthesis and characterization of poly(itaconate-co-citrate-co-octanediol) (PICO) is presented, which exhibits tunable formation of elastomeric networks through radical crosslinking of itaconate in the polymer backbone of viscous polyester gels. Through variation of reaction times and monomer molar composition, materials with modulation of a wide range of elasticity (36-1476 kPa) are generated, indicating the tunability of materials to specific elastomeric constructs. This correlated with measured rapid and controllable gelation times. As a proof of principle, scaffold support for cardiac tissue patches is developed, which presents visible tissue organization and viability with appropriate elastomeric support from PICO materials. These formulations present potential application in a range of healthcare applications with requirement for elastomeric support with controllable, rapid gelation under mild conditions.
Collapse
Affiliation(s)
- Locke Davenport Huyer
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| | - A. Dawn Bannerman
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| | - Yufeng Wang
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| | - Houman Savoji
- Institute of Biomaterials and Biomedical Engineering University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| | - Ericka J. Knee‐Walden
- Institute of Biomaterials and Biomedical Engineering University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| | - Amanda Brissenden
- Department of Chemical Engineering Queen's University Kingston Ontario Canada
| | - Bess Yee
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| | - Mohammad Shoaib
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
| | - Erin Bobicki
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
| | - Brian G. Amsden
- Department of Chemical Engineering Queen's University Kingston Ontario Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto Ontario Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto Toronto Ontario Canada
- Toronto General Research Institute University Health Network Toronto Ontario Canada
| |
Collapse
|
46
|
Nguyen AH, Marsh P, Schmiess-Heine L, Burke PJ, Lee A, Lee J, Cao H. Cardiac tissue engineering: state-of-the-art methods and outlook. J Biol Eng 2019; 13:57. [PMID: 31297148 PMCID: PMC6599291 DOI: 10.1186/s13036-019-0185-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to assess the state-of-the-art fabrication methods, advances in genome editing, and the use of machine learning to shape the prospective growth in cardiac tissue engineering. Those interdisciplinary emerging innovations would move forward basic research in this field and their clinical applications. The long-entrenched challenges in this field could be addressed by novel 3-dimensional (3D) scaffold substrates for cardiomyocyte (CM) growth and maturation. Stem cell-based therapy through genome editing techniques can repair gene mutation, control better maturation of CMs or even reveal its molecular clock. Finally, machine learning and precision control for improvements of the construct fabrication process and optimization in tissue-specific clonal selections with an outlook of cardiac tissue engineering are also presented.
Collapse
Affiliation(s)
- Anh H. Nguyen
- Electrical and Computer Engineering Department, University of Alberta, Edmonton, Alberta Canada
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Paul Marsh
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Lauren Schmiess-Heine
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
| | - Peter J. Burke
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Chemical Engineering and Materials Science Department, University of California Irvine, Irvine, CA USA
| | - Abraham Lee
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Mechanical and Aerospace Engineering Department, University of California Irvine, Irvine, CA USA
| | - Juhyun Lee
- Bioengineering Department, University of Texas at Arlington, Arlington, TX USA
| | - Hung Cao
- Electrical Engineering and Computer Science Department, University of California Irvine, Irvine, CA USA
- Biomedical Engineering Department, University of California Irvine, Irvine, CA USA
- Henry Samueli School of Engineering, University of California, Irvine, USA
| |
Collapse
|
47
|
Microfabrication of AngioChip, a biodegradable polymer scaffold with microfluidic vasculature. Nat Protoc 2019; 13:1793-1813. [PMID: 30072724 DOI: 10.1038/s41596-018-0015-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Microengineered biomimetic systems for organ-on-a-chip or tissue engineering purposes often fail as a result of an inability to recapitulate the in vivo environment, specifically the presence of a well-defined vascular system. To address this limitation, we developed an alternative method to cultivate three-dimensional (3D) tissues by incorporating a microfabricated scaffold, termed AngioChip, with a built-in perfusable vascular network. Here, we provide a detailed protocol for fabricating the AngioChip scaffold, populating it with endothelial cells and parenchymal tissues, and applying it in organ-on-a-chip drug testing in vitro and surgical vascular anastomosis in vivo. The fabrication of the AngioChip scaffold is achieved by a 3D stamping technique, in which an intricate microchannel network can be embedded within a 3D scaffold. To develop a vascularized tissue, endothelial cells are cultured in the lumen of the AngioChip network, and parenchymal cells are encapsulated in hydrogels that are amenable to remodeling around the vascular network to form functional tissues. Together, these steps yield a functional, vascularized network in vitro over a 14-d period. Finally, we demonstrate the functionality of AngioChip-vascularized hepatic and cardiac tissues, and describe direct surgical anastomosis of the AngioChip vascular network on the hind limb of a Lewis rat model.
Collapse
|
48
|
Firoozi N, Kang Y. A Highly Elastic and Autofluorescent Poly(xylitol-dodecanedioic Acid) for Tissue Engineering. ACS Biomater Sci Eng 2019; 5:1257-1267. [PMID: 33405644 DOI: 10.1021/acsbiomaterials.9b00059] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In spite of the vast research on developing a highly elastic polymer for tissue regeneration, using a renewable resource and a simple, environment-friendly synthesis route to synthesize an elastic polymer has not been successfully achieved yet. The objective of this study was to use a simple melt condensation polymerization method to develop an elastic polymer for tissue regeneration applications. A nature-derived renewable, nontoxic, and inexpensive monomer, xylitol, and a cross-linking agent, dodecanedioic acid, were used to synthesize the new polymer named poly(xylitol-dodecanedioic acid) (PXDDA). Its physicochemical and biological properties were fully characterized. Fourier transform infrared (FTIR) results confirmed the formation of ester bonding in the polymer structure, and thermal analysis results demonstrated that the polymer was completely amorphous. The polymer is highly elastic. Increasing the molar ratio of dodecanedioic acid resulted in lower elasticity, higher hydrophobicity, and lower glass transition temperature. Further, the polymer degradation rate and in vitro dye release from the polymer also became slower when the amount of dodecanedioic acid in the composite increased. Biocompatibility studies showed that both the polymeric materials and the degraded products of the polymer did not show any toxicity. Instead, this new polymer significantly promoted cell adhesion and proliferation, compared to a widely used polymer, poly(lactic acid), and tissue culture plates. Interestingly, the PXDDA polymer demonstrated autofluorescent properties. Overall, these results suggest that a new, elastic, biodegradable polymer has been successfully synthesized, and it holds great promise for biomedical applications in drug delivery and tissue engineering.
Collapse
Affiliation(s)
- Negar Firoozi
- Department of Ocean & Mechanical Engineering, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida 33431, United States
| | - Yunqing Kang
- Department of Ocean & Mechanical Engineering, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida 33431, United States.,Department of Biomedical Science, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida 33431, United States.,Integrative Biology Ph.D. Program, Department of Biological Science, Florida Atlantic University, 777 Glades Road, Boca Raton, Florida 33431, United States
| |
Collapse
|
49
|
Boutry CM, Beker L, Kaizawa Y, Vassos C, Tran H, Hinckley AC, Pfattner R, Niu S, Li J, Claverie J, Wang Z, Chang J, Fox PM, Bao Z. Biodegradable and flexible arterial-pulse sensor for the wireless monitoring of blood flow. Nat Biomed Eng 2019; 3:47-57. [DOI: 10.1038/s41551-018-0336-5] [Citation(s) in RCA: 371] [Impact Index Per Article: 74.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
|
50
|
Jang J, Park H, Jeong H, Mo E, Kim Y, Yuk JS, Choi SQ, Kim YW, Shin J. Thermoset elastomers covalently crosslinked by hard nanodomains of triblock copolymers derived from carvomenthide and lactide: tunable strength and hydrolytic degradability. Polym Chem 2019. [DOI: 10.1039/c8py01765d] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sustainable, mechanically reinforced, and hydrolytically degradable thermoset elastomers were synthesized by one-pot, three-step synthesis & crosslinking.
Collapse
Affiliation(s)
- Jeongmin Jang
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Chemical Engineering
| | - Hyejin Park
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Chemical Engineering
| | - Haemin Jeong
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Advanced Materials & Chemical Engineering
| | - Eunbi Mo
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Chemical Engineering
| | - Yongbin Kim
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Chemical Engineering
| | - Jeong Suk Yuk
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
| | - Siyoung Q. Choi
- Department of Chemical and Biomolecular Engineering
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon
- Korea
| | - Young-Wun Kim
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Advanced Materials & Chemical Engineering
| | - Jihoon Shin
- Center for Environment & Sustainable Resources
- Korea Research Institute of Chemical Technology (KRICT)
- Daejeon
- Korea
- Department of Advanced Materials & Chemical Engineering
| |
Collapse
|