1
|
Werschler N, Quintard C, Nguyen S, Penninger J. Engineering next generation vascularized organoids. Atherosclerosis 2024; 398:118529. [PMID: 39304390 DOI: 10.1016/j.atherosclerosis.2024.118529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 09/22/2024]
Abstract
Organoids are self-organizing 3D cell culture models that are valuable for studying the mechanisms underlying both development and disease in multiple species, particularly, in humans. These 3D engineered tissues can mimic the structure and function of human organs in vitro. Methods to generate organoids have substantially improved to better resemble, in various ways, their in vivo counterpart. One of the major limitations in current organoid models is the lack of a functional vascular compartment. Here we discuss methodological approaches to generating perfusable blood vessel networks in organoid systems. Inclusion of perfused vascular compartments markedly enhances the physiological relevance of organoid systems and is a critical step in the establishment of next generation, higher-complexity in vitro systems for use in developmental, clinical, and drug-development settings.
Collapse
Affiliation(s)
- Nicolas Werschler
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, School of Biomedical Engineering, Vancouver, Canada.
| | - Clement Quintard
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, Medical Genetics, Vancouver, Canada
| | - Stephanie Nguyen
- University of British Columbia, School of Biomedical Engineering, Vancouver, Canada
| | - Josef Penninger
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, School of Biomedical Engineering, Vancouver, Canada; University of British Columbia, Medical Genetics, Vancouver, Canada; Helmholtz Centre for Infection Research, Germany; Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Austria; IMBA Institute of Molecular Biotechnology, Vienna, Austria
| |
Collapse
|
2
|
Dong Y, Zhou X, Ding Y, Luo Y, Zhao H. Advances in tumor microenvironment: Applications and challenges of 3D bioprinting. Biochem Biophys Res Commun 2024; 730:150339. [PMID: 39032359 DOI: 10.1016/j.bbrc.2024.150339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
The tumor microenvironment (TME) assumes a pivotal role in the treatment of oncological diseases, given its intricate interplay of diverse cellular components and extracellular matrices. This dynamic ecosystem poses a serious challenge to traditional research methods in many ways, such as high research costs, inefficient translation, poor reproducibility, and low modeling success rates. These challenges require the search for more suitable research methods to accurately model the TME, and the emergence of 3D bioprinting technology is transformative and an important complement to these traditional methods to precisely control the distribution of cells, biomolecules, and matrix scaffolds within the TME. Leveraging digital design, the technology enables personalized studies with high precision, providing essential experimental flexibility. Serving as a critical bridge between in vitro and in vivo studies, 3D bioprinting facilitates the realistic 3D culturing of cancer cells. This comprehensive article delves into cutting-edge developments in 3D bioprinting, encompassing diverse methodologies, biomaterial choices, and various 3D tumor models. Exploration of current challenges, including limited biomaterial options, printing accuracy constraints, low reproducibility, and ethical considerations, contributes to a nuanced understanding. Despite these challenges, the technology holds immense potential for simulating tumor tissues, propelling personalized medicine, and constructing high-resolution organ models, marking a transformative trajectory in oncological research.
Collapse
Affiliation(s)
- Yingying Dong
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xue Zhou
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Yunyi Ding
- Department of Emergency Medicine, The Second Affiliated Hospital of Zhejiang University, School, Hangzhou, 310009, China.
| | - Yichen Luo
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Hong Zhao
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China; Department of Breast Surgery, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310060, China.
| |
Collapse
|
3
|
Fan R, Wu J, Duan S, Jin L, Zhang H, Zhang C, Zheng A. Droplet-based microfluidics for drug delivery applications. Int J Pharm 2024; 663:124551. [PMID: 39106935 DOI: 10.1016/j.ijpharm.2024.124551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
The microfluidic method primainly utilizes two incompatible liquids as continuous phase and dispersed phase respectively. It controls the formation of droplets by managing the microchannel structure and the flow rate ratio of the two phases. Droplet-based microfluidics is a rapidly expanding interdisciplinary research field encompassing physics, biochemistry, and Microsystems engineering. Droplet microfluidics offer a diverse and practical toolset that enables chemical and biological experiments to be conducted at high speeds and with greater efficiency compared to traditional instruments. The applications of droplet-based microfluidics are vast, including areas such as drug delivery, owing to its compatibility with numerous chemical and biological reagents and its ability to carry out various operations. This technology has been extensively researched due to its promising features. In this review, we delve into the materials used in droplet generation-based microfluidic devices, manufacturing techniques, methods for droplet generation in channels, and, finally, we summarize the applications of droplet generation-based microfluidics in drug delivery vectors, encompassing nanoparticles, microspheres, microcapsules, and hydrogel particles. We also discuss the challenges and future prospects of this technology across a wide array of applications.
Collapse
Affiliation(s)
- Ranran Fan
- College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Jie Wu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Shuwei Duan
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Lili Jin
- College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin Province 133002, China
| | - Hui Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Changhao Zhang
- College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University College of Pharmacy, Yanji, Jilin Province 133002, China.
| | - Aiping Zheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
4
|
López-León CF, Planet R, Soriano J. Preparation and Mechano-Functional Characterization of PEGylated Fibrin Hydrogels: Impact of Thrombin Concentration. Gels 2024; 10:116. [PMID: 38391447 PMCID: PMC10888336 DOI: 10.3390/gels10020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Three-dimensional (3D) neuronal cultures grown in hydrogels are promising platforms to design brain-like neuronal networks in vitro. However, the optimal properties of such cultures must be tuned to ensure a hydrogel matrix sufficiently porous to promote healthy development but also sufficiently rigid for structural support. Such an optimization is difficult since it implies the exploration of different hydrogel compositions and, at the same time, a functional analysis to validate neuronal culture viability. To advance in this quest, here we present a combination of a rheological protocol and a network-based functional analysis to investigate PEGylated fibrin hydrogel networks with gradually higher stiffness, achieved by increasing the concentration of thrombin. We observed that moderate thrombin concentrations of 10% and 25% in volume shaped healthy networks, although the functional traits depended on the hydrogel stiffness, which was much higher for the latter concentration. Thrombin concentrations of 65% or higher led to networks that did not survive. Our results illustrate the difficulties and limitations in preparing 3D neuronal networks, and stress the importance of combining a mechano-structural characterization of a biomaterial with a functional one.
Collapse
Affiliation(s)
- Clara F López-León
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelon, E-08028 Barcelona, Spain
| | - Ramon Planet
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelon, E-08028 Barcelona, Spain
| | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), Universitat de Barcelon, E-08028 Barcelona, Spain
| |
Collapse
|
5
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Cheng KC, Theato P, Hsu SH. 3D-bioprintable endothelial cell-laden sacrificial ink for fabrication of microvessel networks. Biofabrication 2023; 15:045026. [PMID: 37722376 DOI: 10.1088/1758-5090/acfac1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 09/20/2023]
Abstract
Although various research efforts have been made to produce a vascular-like network structure as scaffolds for tissue engineering, there are still several limitations. Meanwhile, no articles have been published on the direct embedding of cells within a glucose sensitive sacrificial hydrogel followed by three-dimensional (3D) bioprinting to fabricate vascular structures. In this study, the hydrogel composed of reversibly crosslinked poly(ethylene glycol) diacrylate and dithiothreitol with borax and branched polyethylenimine was used as the sacrificial hydrogel to fabricate vascular-like network structure. The component proportion ratio of the sacrificial hydrogel was optimized to achieve proper self-healing, injectable, glucose-sensitive, and 3D printing properties through the balance of boronate ester bond, hydrogen bond, and steric hinderance effect. The endothelial cells (ECs) can be directly embedded into sacrificial hydrogel and then bioprinted through a 110μm nozzle into the neural stem cell (NSC)-laden non-sacrificial hydrogel, forming the customized EC-laden vascularized microchannel (one-step). The EC-laden sacrificial hydrogel was dissolved immediately in the medium while cells kept growing. The ECs proliferated well within the vascularized microchannel structure and were able to migrate to the non-sacrificial hydrogel in one day. ECs and NSCs interacted around the vascularized microchannel to form capillary-like structure and vascular-like structure expressing CD31 in 14 d. The sacrificial hydrogel conveniently prepared from commercially available chemicals through simple mixing can be used in 3D bioprinting to create customized and complex but easily removable vascularized structure for tissue engineering applications.
Collapse
Affiliation(s)
- Kun-Chih Cheng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Patrick Theato
- Institute for Chemical Technology and Polymer Chemistry, Karlsruhe Institute of Technology (KIT), Engesser Str. 18, D-76131 Karlsruhe, Germany
- Institute for Biological Interfaces III (IBG3), Hermann-von-Helmholtz-Platz 1, D-76344 Eggenstein-Leopoldshafen, Germany
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| |
Collapse
|
7
|
Li X, Coates DE. Hollow channels scaffold in bone regenerative: a review. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:1702-1715. [PMID: 36794303 DOI: 10.1080/09205063.2023.2181066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023]
Abstract
Bone substitute materials have been extensively used for bone regeneration over the past 50 years. The development of novel materials, fabrication technologies and the incorporation and release of regenerative cytokines, growth factors, cells and antimicrobials has been driven by the rapid development in the field of additive manufacturing technology. There are still however, significant challenges that need addressing, including ways to better mediate the rapid vascularization of bone scaffolds to enhance subsequent regeneration and osteogenesis. Increasing construct porosity can accelerate the development of blood vessels in the scaffold, but doing so also weakens the constructs mechanical properties. A novel design for promoting rapid vascularization is to fabricate custom-made hollow channels as bone scaffolds. Summarized here are the current developments in hollow channels scaffold, including their biological attributes, physio-chemical properties, and effects on regeneration. An overview of recent developments in scaffold fabrication as they relate to hollow channel constructs and their structural features will be introduced with an emphasis on attributes that enhance new bone and vessel formation. Furthermore, the potential to enhance angiogenesis and osteogenesis by replicating the structure of real bone will be highlighted.
Collapse
Affiliation(s)
- Xiao Li
- University of Otago, Dunedin, New Zealand
| | - Dawn Elizabeth Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
8
|
In vitro cell stretching devices and their applications: From cardiomyogenic differentiation to tissue engineering. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2023. [DOI: 10.1016/j.medntd.2023.100220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
9
|
Bi W, Cai S, Lei T, Wang L. Implementation of blood-brain barrier on microfluidic chip: recent advance and future prospects. Ageing Res Rev 2023; 87:101921. [PMID: 37004842 DOI: 10.1016/j.arr.2023.101921] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/02/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
The complex structure of the blood-brain barrier (BBB) hinders its modeling and the treatment of brain diseases. The microfluidic technology promotes the development of BBB-on-a-chip platforms, which can be used to reproduce the complex brain microenvironment and physiological reactions. Compared with traditional transwell technology, microfluidic BBB-on-a-chip shows great technical advantages in terms of flexible control of fluid shear stress in the chip and fabrication efficiency of the chip system, which can be enhanced by the development of lithography and three-dimensional (3D) printing. It is convenient to accurately monitor the dynamic changes of biochemical parameters of individual cells in the model by integrating an automatic super-resolution imaging sensing platform. In addition, biomaterials, especially hydrogels and conductive polymers, solve the limitations of microfluidic BBB-on-a-chip by compounding onto microfluidic chip to provide a 3D space and special performance on the microfluidic chip. The microfluidic BBB-on-a-chip promotes the development of basic research, including cell migration, mechanism exploration of neurodegenerative diseases, drug barrier permeability, SARS-CoV-2 pathology. This study summarizes the recent advances, challenges and future prospects of microfluidic BBB-on-a-chip, which can help to promote the development of personalized medicine and drug discovery.
Collapse
|
10
|
Tolabi H, Davari N, Khajehmohammadi M, Malektaj H, Nazemi K, Vahedi S, Ghalandari B, Reis RL, Ghorbani F, Oliveira JM. Progress of Microfluidic Hydrogel-Based Scaffolds and Organ-on-Chips for the Cartilage Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2208852. [PMID: 36633376 DOI: 10.1002/adma.202208852] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/09/2022] [Indexed: 05/09/2023]
Abstract
Cartilage degeneration is among the fundamental reasons behind disability and pain across the globe. Numerous approaches have been employed to treat cartilage diseases. Nevertheless, none have shown acceptable outcomes in the long run. In this regard, the convergence of tissue engineering and microfabrication principles can allow developing more advanced microfluidic technologies, thus offering attractive alternatives to current treatments and traditional constructs used in tissue engineering applications. Herein, the current developments involving microfluidic hydrogel-based scaffolds, promising structures for cartilage regeneration, ranging from hydrogels with microfluidic channels to hydrogels prepared by the microfluidic devices, that enable therapeutic delivery of cells, drugs, and growth factors, as well as cartilage-related organ-on-chips are reviewed. Thereafter, cartilage anatomy and types of damages, and present treatment options are briefly overviewed. Various hydrogels are introduced, and the advantages of microfluidic hydrogel-based scaffolds over traditional hydrogels are thoroughly discussed. Furthermore, available technologies for fabricating microfluidic hydrogel-based scaffolds and microfluidic chips are presented. The preclinical and clinical applications of microfluidic hydrogel-based scaffolds in cartilage regeneration and the development of cartilage-related microfluidic chips over time are further explained. The current developments, recent key challenges, and attractive prospects that should be considered so as to develop microfluidic systems in cartilage repair are highlighted.
Collapse
Affiliation(s)
- Hamidreza Tolabi
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran, 15875-4413, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 15875-4413, Iran
| | - Niyousha Davari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
| | - Mehran Khajehmohammadi
- Department of Mechanical Engineering, Faculty of Engineering, Yazd University, Yazd, 89195-741, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 8916877391, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg, 9220, Denmark
| | - Katayoun Nazemi
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Samaneh Vahedi
- Department of Material Science and Engineering, Faculty of Engineering, Imam Khomeini International University, Qazvin, 34149-16818, Iran
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| | - Farnaz Ghorbani
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058, Erlangen, Germany
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| |
Collapse
|
11
|
In Vitro and In Vivo Investigation on the Effectiveness of Alginate-Based Gastric Mucosal Protective Gel. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8287163. [PMID: 36060134 PMCID: PMC9433266 DOI: 10.1155/2022/8287163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 07/08/2022] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
Objective To investigate the feasibility and effectiveness of an alginate-based gastric mucosal protective gel on the gastric ulcer. Methods (1) In the physical protection model, after GES-1 cell attachment add the gel to transwell chamber, add different concentrations of HCl to the gel. Absorbance was measured to assess proliferation and images of the cells migrating into the wound were taken; then the migration rate of the cells was quantified by comparing images. (2) In the gastric ulcer model, excise the gastric mucosal of SD rats; the gel and fixative were applied on the artificial ulcer immediately. Dissect rats after 10 days, and calculate the wound healing rate and analyzed histology changes. Results The effect of hydrochloric acid on cells in the lower layer was significantly reduced after the use of gastric mucosal protection gel. The protective gel had an isolation effect on different concentrations of acid. A number of GES-1 were significantly higher than those in the control group at 24 h to 72 h (P < 0.01). The migration was observed compared with the control group. The average healing rate of ulcer in the gel group was about 50%, and the control group was about 30%. Inflammation occurred in all wound regions after ten days. In the gel group, inflammatory infiltration depth was lower than that of the control, and part of SD rats' new muscle layer appeared without inflammatory infiltration. The connective tissue proliferation promoted tissue repair. In the control group, necrosis marginal, mucosal hyperplasia, marginal lymphocyte aggregation, and bleeding were observed. Conclusion This novel gel mainly has an isolating and shielding effect to prevent the wound from being exposed to gastric acid for a long time, and it can reduce the inflammatory reaction on the wounds to promote the healing of the ulcer. The gastric mucosal protective gel cannot only promote the speed of wound healing but also improve the quality of wound healing.
Collapse
|
12
|
Hospodiuk-Karwowski M, Chi K, Pritchard J, Catchmark JM. Vascularized pancreas-on-a-chip device produced using a printable simulated extracellular matrix. Biomed Mater 2022; 17. [PMID: 36001993 DOI: 10.1088/1748-605x/ac8c74] [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: 04/20/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022]
Abstract
The extracellular matrix (ECM) influences cellular behavior, function, and fate. The ECM surrounding Langerhans islets has not been investigated in detail to explain its role in the development and maturation of pancreatic β-cells. Herein, a complex combination of the simulated ECM (sECM) has been examined with a comprehensive analysis of cell response and a variety of controls. The most promising results were obtained from group containing fibrin, collagen type I, Matrigel®, hyaluronic acid, methylcellulose, and two compounds of functionalized, ionically crosslinking bacterial cellulose (sECMbc). Even though the cell viability was not significantly impacted, the performance of group of sECMbc showed 2 to 4x higher sprouting number and length, 2 to 4x higher insulin secretion in static conditions, and 2 to 10x higher gene expression of VEGF-A, Endothelin-1, and NOS3 than the control group of fibrin matrix (sECMf). Each material was tested in a hydrogel-based, perfusable, pancreas-on-a-chip device and the best group - sECMbc has been tested with the drug Sunitinib to show the extended possibilities of the device for both diabetes-like screening as well as PDAC chemotherapeutics screening for potential personal medicine approach. It proved its functionality in 7 days dynamic culture and is suitable as a physiological tissue model. Moreover, the device with the pancreatic-like spheroids was 3D bioprintable and perfusable.
Collapse
Affiliation(s)
- Monika Hospodiuk-Karwowski
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Kai Chi
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Justin Pritchard
- Biomedical Engineering Department, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Jeffrey M Catchmark
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| |
Collapse
|
13
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
14
|
Mei J, Vasan A, Magaram U, Takemura K, Chalasani SH, Friend J. Well-free agglomeration and on-demand three-dimensional cell cluster formation using guided surface acoustic waves through a couplant layer. Biomed Microdevices 2022; 24:18. [PMID: 35596837 PMCID: PMC9124176 DOI: 10.1007/s10544-022-00617-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2022] [Indexed: 11/24/2022]
Abstract
Three-dimensional cell agglomerates are broadly useful in tissue engineering and drug testing. We report a well-free method to form large (1.4-mm) multicellular clusters using 100-MHz surface acoustic waves (SAW) without direct contact with the media or cells. A fluid couplant is used to transform the SAW into acoustic streaming in the cell-laden media held in a petri dish. The couplant transmits longitudinal sound waves, forming a Lamb wave in the petri dish that, in turn, produces longitudinal sound in the media. Due to recirculation, human embryonic kidney (HEK293) cells in the dish are carried to the center of the coupling location, forming a cluster in less than 10 min. A few minutes later, these clusters may then be translated and merged to form large agglomerations, and even repeatedly folded to produce a roughly spherical shape of over 1.4 mm in diameter for incubation-without damaging the existing intercellular bonds. Calcium ion signaling through these clusters and confocal images of multiprotein junctional complexes suggest a continuous tissue construct: intercellular communication. They may be formed at will, and the method is feasibly useful for formation of numerous agglomerates in a single petri dish.
Collapse
Affiliation(s)
- Jiyang Mei
- Medically Advanced Devices Laboratory, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Dr MC0411, La Jolla, San Diego, CA, 92093, USA
| | - Aditya Vasan
- Medically Advanced Devices Laboratory, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Dr MC0411, La Jolla, San Diego, CA, 92093, USA
| | - Uri Magaram
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, San Diego, CA, 92037, USA
| | - Kenjiro Takemura
- Department of Mechanical Engineering, Keio University, 3-14-1 Hiyoshi, Kouhoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Sreekanth H Chalasani
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd, La Jolla, San Diego, CA, 92037, USA
| | - James Friend
- Medically Advanced Devices Laboratory, Department of Mechanical and Aerospace Engineering, Jacobs School of Engineering and Department of Surgery, School of Medicine, University of California San Diego, 9500 Gilman Dr MC0411, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
15
|
Bjørge IM, Correia CR, Mano JF. Hipster microcarriers: exploring geometrical and topographical cues of non-spherical microcarriers in biomedical applications. MATERIALS HORIZONS 2022; 9:908-933. [PMID: 34908074 DOI: 10.1039/d1mh01694f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Structure and organisation are key aspects of the native tissue environment, which ultimately condition cell fate via a myriad of processes, including the activation of mechanotransduction pathways. By modulating the formation of integrin-mediated adhesions and consequently impacting cell contractility, engineered geometrical and topographical cues may be introduced to activate downstream signalling and ultimately control cell morphology, proliferation, and differentiation. Microcarriers appear as attractive vehicles for cell-based tissue engineering strategies aiming to modulate this 3D environment, but also as vehicles for cell-free applications, given the ease in tuning their chemical and physical properties. In this review, geometry and topography are highlighted as two preponderant features in actively regulating interactions between cells and the extracellular matrix. While most studies focus on the 2D environment, we focus on how the incorporation of these strategies in 3D systems could be beneficial. The techniques applied to design 3D microcarriers with unique geometries and surface topographical cues are covered, as well as specific tissue engineering approaches employing these microcarriers. In fact, successfully achieving a functional histoarchitecture may depend on a combination of fine-tuned geometrically shaped microcarriers presenting intricately tailored topographical cues. Lastly, we pinpoint microcarrier geometry as a key player in cell-free biomaterial-based strategies, and its impact on drug release kinetics, the production of steerable microcarriers to target tumour cells, and as protein or antibody biosensors.
Collapse
Affiliation(s)
- Isabel M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - Clara R Correia
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
16
|
Wang X, Li S, Yu H, Lv J, Fan M, Wang X, Wang X, Liang Y, Mao L, Zhao Z. The Biocompatibility of Multi-Source Stem Cells and Gelatin-Carboxymethyl Chitosan-Sodium Alginate Hybrid Biomaterials. Tissue Eng Regen Med 2022; 19:491-503. [PMID: 35119649 PMCID: PMC9130400 DOI: 10.1007/s13770-021-00429-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nowadays, biological tissue engineering is a growing field of research. Biocompatibility is a key indicator for measuring tissue engineering biomaterials, which is of great significance for the replacement and repair of damaged tissues. METHODS In this study, using gelatin, carboxymethyl chitosan, and sodium alginate, a tissue engineering material scaffold that can carry cells was successfully prepared. The material was characterized by Fourier transforms infrared spectroscopy. In addition, the prepared scaffolds have physicochemical properties, such as swelling ratio, biodegradability. we observed the biocompatibility of the hydrogel to different adult stem cells (BMSCs and ADSCs) in vivo and in vitro. Adult stem cells were planted on gelatin-carboxymethyl chitosan-sodium alginate (Gel/SA/CMCS) hydrogels for 7 days in vitro, and the survival of stem cells in vitro was observed by live/died staining. Gel/SA/CMCS hydrogels loaded with stem cells were subcutaneously transplanted into nude mice for 14 days of in vivo culture observation. The survival of adult stem cells was observed by staining for stem cell surface markers (CD29, CD90) and Ki67. RESULTS The scaffolds had a microporous structure with an appropriate pore size (about 80 μm). Live/died staining showed that adult stem cells could stably survive in Gel/SA/CMCS hydrogels for at least 7 days. After 14 days of culture in nude mice, Ki67 staining showed that the stem cells supported by Gel/SA/CMCS hydrogel still had high proliferation activity. CONCLUSION Gel/SA/CMCSs hydrogel has a stable interpenetrating porous structure, suitable swelling performance and degradation rate, can promote and support the survival of adult stem cells in vivo and in vitro, and has good biocompatibility. Therefore, Gel/SA/CMCS hydrogel is a strong candidate for biological tissue engineering materials.
Collapse
Affiliation(s)
- Xinzhe Wang
- Clinical Medical College, Jining Medical University, Jining, 272067 Shandong China
| | - Siqi Li
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272067 Shandong China
| | - Honglian Yu
- Department of Biochemistry, Jining Medical University, Jining, Shandong 272067 People’s Republic of China ,Collaborative Innovation Center, Jining Medical University, Jining, Shandong 272067 People’s Republic of China
| | - Jianzhi Lv
- Clinical Medical College, Jining Medical University, Jining, 272067 Shandong China
| | - Minglun Fan
- Clinical Medical College, Jining Medical University, Jining, 272067 Shandong China
| | - Ximing Wang
- Clinical Medical College, Jining Medical University, Jining, 272067 Shandong China
| | - Xin Wang
- Clinical Medical College, Jining Medical University, Jining, 272067 Shandong China
| | - Yanting Liang
- Basic Medicine College, Jining Medical University, Jining, Shandong 272067 People’s Republic of China
| | - Lingna Mao
- Basic Medicine College, Jining Medical University, Jining, Shandong 272067 People’s Republic of China
| | - Zhankui Zhao
- Department of Urology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272100 People’s Republic of China
| |
Collapse
|
17
|
Kumari G, Abhishek K, Singh S, Hussain A, Altamimi MA, Madhyastha H, Webster TJ, Dev A. A voyage from 3D to 4D printing in nanomedicine and healthcare: part II. Nanomedicine (Lond) 2022; 17:255-270. [PMID: 35109687 DOI: 10.2217/nnm-2021-0454] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Recent advancements in biomedical tissue engineering are gaining wide interest. Implementing biology of living cells and organisms using technological solutions such as incorporating 4D printing and bioprinting for tissue regeneration/tissue repair, organ regeneration, early diagnosis of deadly diseases (particularly cancer, cardiac disorders and tuberculosis) has successfully opened a new generation of biomedical research. The present review primarily addresses the clinical application of 4D printing and bioprinting techniques for applications such as early detection of diseases and drug delivery. Notably, this review continues the discussion from part I regarding published informative data, in vitro and in vivo findings, commercial biosensors for early disease diagnosis, drug delivery and current challenges in 4D printing/bioprinting.
Collapse
Affiliation(s)
- Gourvi Kumari
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Kumar Abhishek
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Sneha Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Afzal Hussain
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Mohammad A Altamimi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, School of Medicine, University of Miyazaki, Miyazaki, 889 1692, Japan
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| |
Collapse
|
18
|
Tarrahi R, Khataee A, Karimi A, Yoon Y. The latest achievements in plant cellulose-based biomaterials for tissue engineering focusing on skin repair. CHEMOSPHERE 2022; 288:132529. [PMID: 34637866 DOI: 10.1016/j.chemosphere.2021.132529] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/27/2021] [Accepted: 10/08/2021] [Indexed: 06/13/2023]
Abstract
The present work reviews recent developments in plant cellulose-based biomaterial design and applications, properties, characterizations, and synthesis for skin tissue engineering and wound healing. Cellulose-based biomaterials are promising materials for their remarkable adaptability with three-dimensional polymeric structure. They are capable of mimicking tissue properties, which plays a key role in tissue engineering. Besides, concerns for environmental issues have motivated scientists to move toward eco-friendly materials and natural polymer-based materials for applications in the tissue engineering field these days. Therefore, cellulose as an appropriate substitute for common polymers based on crude coal, animal, and human-derived biomolecules is greatly considered for various applications in biomedical fields. Generally, natural biomaterials lack good mechanical properties for skin tissue engineering. But using modified cellulose-based biopolymers tackles these restrictions and prevents immunogenic responses. Moreover, tissue engineering is a quick promoting field focusing on the generation of novel biomaterials with modified characteristics to improve scaffold function through physical, biochemical, and chemical tailoring. Also, nanocellulose with a broad range of applications, particularly in tissue engineering, advanced wound dressing, and as a material for coupling with drugs and sensorics, has been reviewed here. Moreover, the potential cytotoxicity and immunogenicity of cellulose-based biomaterials are addressed in this review.
Collapse
Affiliation(s)
- Roshanak Tarrahi
- Health Promotion Research Center, Iran University of Medical Sciences, 14496-14535, Tehran, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471, Tabriz, Iran; Department of Environmental Engineering, Gebze Technical University, 41400, Gebze, Turkey
| | - Afzal Karimi
- Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, 1449614535, Tehran, Iran
| | - Yeojoon Yoon
- Department of Environmental and Energy Engineering, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
19
|
Mirzaei R, Ranjbar R. Hijacking host components for bacterial biofilm formation: An advanced mechanism. Int Immunopharmacol 2022; 103:108471. [PMID: 34952466 DOI: 10.1016/j.intimp.2021.108471] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
Biofilm is a community of bacteria embedded in the extracellular matrix that accounts for 80% of bacterial infections. Biofilm enables bacterial cells to provide particular conditions and produce virulence determinants in response to the unavailability of micronutrients and local oxygen, resulting in their resistance to various antibacterial agents. Besides, the human immune reactions are not completely competent in the elimination of biofilm. Most importantly, the growing body of evidence shows that some bacterial spp. use a variety of mechanisms by which hijack the host components to form biofilm. In this regard, host components, such as DNA, hyaluronan, collagen, fibronectin, mucin, oligosaccharide moieties, filamentous polymers (F-actin), plasma, platelets, keratin, sialic acid, laminin, vitronectin, C3- and C4- binding proteins, antibody, proteases, factor I, factor H, and acidic proline-rich proteins have been reviewed. Hence, the characterization of interactions between bacterial biofilm and the host would be critical to effectively address biofilm-associated infections. In this paper, we review the latest information on the hijacking of host factors by bacteria to form biofilm.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Mohajeri M, Eskandari M, Ghazali ZS, Ghazali HS. Cell encapsulation in alginate-based microgels using droplet microfluidics; a review on gelation methods and applications. Biomed Phys Eng Express 2022; 8. [PMID: 35073537 DOI: 10.1088/2057-1976/ac4e2d] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/24/2022] [Indexed: 11/12/2022]
Abstract
Cell encapsulation within the microspheres using a semi-permeable polymer allows the two-way transfer of molecules such as oxygen, nutrients, and growth factors. The main advantages of cell encapsulation technology include controlling the problems involved in transplanting rejection in tissue engineering applications and reducing the long-term need for immunosuppressive drugs following organ transplantation to eliminate the side effects. Cell-laden microgels can also be used in 3D cell cultures, wound healing, and cancerous clusters for drug testing. Since cell encapsulation is used for different purposes, several techniques have been developed to encapsulate cells. Droplet-based microfluidics is one of the most valuable techniques in cell encapsulating. This study aimed to review the geometries and the mechanisms proposed in microfluidic systems to precisely control cell-laden microgels production with different biopolymers. We also focused on alginate gelation techniques due to their essential role in cell encapsulation applications. Finally, some applications of these microgels and researches will be explored.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Biomedical Engineering Department, Amirkabir University of Technology, Department of Biomedical Engineering No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Mahnaz Eskandari
- Biomedical Engineering Department, Amirkabir University of Technology, Department of Biomedical Engineering No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Zahra Sadat Ghazali
- Biomedical Engineering Department, Amirkabir University of Technology, No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Hanieh Sadat Ghazali
- Department of Nanobiotechnology, Tarbiat Modares University, Jalal Aleahmad-Tehran-Iran, Tehran, 14115-111, Iran (the Islamic Republic of)
| |
Collapse
|
21
|
Tong A, Voronov R. A Minireview of Microfluidic Scaffold Materials in Tissue Engineering. Front Mol Biosci 2022; 8:783268. [PMID: 35087865 PMCID: PMC8787357 DOI: 10.3389/fmolb.2021.783268] [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: 09/25/2021] [Accepted: 12/14/2021] [Indexed: 01/09/2023] Open
Abstract
In 2020, nearly 107,000 people in the U.S needed a lifesaving organ transplant, but due to a limited number of donors, only ∼35% of them have actually received it. Thus, successful bio-manufacturing of artificial tissues and organs is central to satisfying the ever-growing demand for transplants. However, despite decades of tremendous investments in regenerative medicine research and development conventional scaffold technologies have failed to yield viable tissues and organs. Luckily, microfluidic scaffolds hold the promise of overcoming the major challenges associated with generating complex 3D cultures: 1) cell death due to poor metabolite distribution/clearing of waste in thick cultures; 2) sacrificial analysis due to inability to sample the culture non-invasively; 3) product variability due to lack of control over the cell action post-seeding, and 4) adoption barriers associated with having to learn a different culturing protocol for each new product. Namely, their active pore networks provide the ability to perform automated fluid and cell manipulations (e.g., seeding, feeding, probing, clearing waste, delivering drugs, etc.) at targeted locations in-situ. However, challenges remain in developing a biomaterial that would have the appropriate characteristics for such scaffolds. Specifically, it should ideally be: 1) biocompatible-to support cell attachment and growth, 2) biodegradable-to give way to newly formed tissue, 3) flexible-to create microfluidic valves, 4) photo-crosslinkable-to manufacture using light-based 3D printing and 5) transparent-for optical microscopy validation. To that end, this minireview summarizes the latest progress of the biomaterial design, and of the corresponding fabrication method development, for making the microfluidic scaffolds.
Collapse
Affiliation(s)
- Anh Tong
- Otto H. York Department of Chemical and Materials Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Roman Voronov
- Otto H. York Department of Chemical and Materials Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
- Department of Biomedical Engineering, Newark College of Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| |
Collapse
|
22
|
Russo M, Cejas CM, Pitingolo G. Advances in microfluidic 3D cell culture for preclinical drug development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:163-204. [PMID: 35094774 DOI: 10.1016/bs.pmbts.2021.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Drug development is often a very long, costly, and risky process due to the lack of reliability in the preclinical studies. Traditional current preclinical models, mostly based on 2D cell culture and animal testing, are not full representatives of the complex in vivo microenvironments and often fail. In order to reduce the enormous costs, both financial and general well-being, a more predictive preclinical model is needed. In this chapter, we review recent advances in microfluidic 3D cell culture showing how its development has allowed the introduction of in vitro microphysiological systems, laying the foundation for organ-on-a-chip technology. These findings provide the basis for numerous preclinical drug discovery assays, which raise the possibility of using micro-engineered systems as emerging alternatives to traditional models, based on 2D cell culture and animals.
Collapse
Affiliation(s)
- Maria Russo
- Microfluidics, MEMS, Nanostructures (MMN), CNRS UMR 8231, Institut Pierre Gilles de Gennes (IPGG) ESPCI Paris, PSL Research University, Paris France.
| | - Cesare M Cejas
- Microfluidics, MEMS, Nanostructures (MMN), CNRS UMR 8231, Institut Pierre Gilles de Gennes (IPGG) ESPCI Paris, PSL Research University, Paris France
| | - Gabriele Pitingolo
- Bioassays, Microsystems and Optical Engineering Unit, BIOASTER, Paris France
| |
Collapse
|
23
|
Piras CC, Kay AG, Genever PG, Fitremann J, Smith DK. Self-assembled gel tubes, filaments and 3D-printing with in situ metal nanoparticle formation and enhanced stem cell growth. Chem Sci 2022; 13:1972-1981. [PMID: 35308847 PMCID: PMC8848986 DOI: 10.1039/d1sc06062g] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/16/2022] [Indexed: 12/18/2022] Open
Abstract
This paper reports simple strategies to fabricate self-assembled artificial tubular and filamentous systems from a low molecular weight gelator (LMWG). In the first strategy, tubular ‘core–shell’ gel structures based on the dibenzylidenesorbitol-based LMWG DBS-CONHNH2 were made in combination with the polymer gelator (PG) calcium alginate. In the second approach, gel filaments based on DBS-CONHNH2 alone were prepared by wet spinning at elevated concentrations using a ‘solvent-switch’ approach. The higher concentrations used in wet-spinning prevent the need for a supporting PG. Furthermore, this can be extended into a 3D-printing method, with the printed LMWG objects showing excellent stability for at least a week in water. The LMWG retains its unique ability for in situ precious metal reduction, yielding Au nanoparticles (AuNPs) within the tubes and filaments when they are exposed to AuCl3 solutions. Since the gel filaments have a higher loading of DBS-CONHNH2, they can be loaded with significantly more AuNPs. Cytotoxicity and viability studies on human mesenchymal stem cells show that the DBS-CONHNH2 and DBS-CONHNH2/alginate hybrid gels loaded with AuNPs are biocompatible, with the presence of AuNPs enhancing stem cell metabolism. Taken together, these results indicate that DBS-CONHNH2 can be shaped and 3D-printed, and has considerable potential for use in tissue engineering applications. Simple fabrication and 3D-printing methods are used to generate tubes and filaments from self-assembled gels, which can be loaded in situ with gold nanoparticles, with the resulting gels encouraging stem cell proliferation.![]()
Collapse
Affiliation(s)
- Carmen C. Piras
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Alasdair G. Kay
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Paul G. Genever
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Juliette Fitremann
- IMRCP, UMR 5623, CNRS, Université de Toulouse, 118 Route de Narbonne, F-31062 Toulouse, France
| | - David K. Smith
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| |
Collapse
|
24
|
Christov IC. Soft hydraulics: from Newtonian to complex fluid flows through compliant conduits. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2021; 34:063001. [PMID: 34678790 DOI: 10.1088/1361-648x/ac327d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/22/2021] [Indexed: 06/13/2023]
Abstract
Microfluidic devices manufactured from soft polymeric materials have emerged as a paradigm for cheap, disposable and easy-to-prototype fluidic platforms for integrating chemical and biological assays and analyses. The interplay between the flow forces and the inherently compliant conduits of such microfluidic devices requires careful consideration. While mechanical compliance was initially a side-effect of the manufacturing process and materials used, compliance has now become a paradigm, enabling new approaches to microrheological measurements, new modalities of micromixing, and improved sieving of micro- and nano-particles, to name a few applications. This topical review provides an introduction to the physics of these systems. Specifically, the goal of this review is to summarize the recent progress towards a mechanistic understanding of the interaction between non-Newtonian (complex) fluid flows and their deformable confining boundaries. In this context, key experimental results and relevant applications are also explored, hand-in-hand with the fundamental principles for their physics-based modeling. The key topics covered include shear-dependent viscosity of non-Newtonian fluids, hydrodynamic pressure gradients during flow, the elastic response (deformation and bulging) of soft conduits due to flow within, the effect of cross-sectional conduit geometry on the resulting fluid-structure interaction, and key dimensionless groups describing the coupled physics. Open problems and future directions in this nascent field of soft hydraulics, at the intersection of non-Newtonian fluid mechanics, soft matter physics, and microfluidics, are noted.
Collapse
Affiliation(s)
- Ivan C Christov
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, United States of America
| |
Collapse
|
25
|
Shakeri A, Khan S, Didar TF. Conventional and emerging strategies for the fabrication and functionalization of PDMS-based microfluidic devices. LAB ON A CHIP 2021; 21:3053-3075. [PMID: 34286800 DOI: 10.1039/d1lc00288k] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Microfluidics is an emerging and multidisciplinary field that is of great interest to manufacturers in medicine, biotechnology, and chemistry, as it provides unique tools for the development of point-of-care diagnostics, organs-on-chip systems, and biosensors. Polymeric microfluidics, unlike glass and silicon, offer several advantages such as low-cost mass manufacturing and a wide range of beneficial material properties, which make them the material of choice for commercial applications and high-throughput systems. Among polymers used for the fabrication of microfluidic devices, polydimethylsiloxane (PDMS) still remains the most widely used material in academia due to its advantageous properties, such as excellent transparency and biocompatibility. However, commercialization of PDMS has been a challenge mostly due to the high cost of the current fabrication strategies. Moreover, specific surface modification and functionalization steps are required to tailor the surface chemistry of PDMS channels (e.g. biomolecule immobilization, surface hydrophobicity and antifouling properties) with respect to the desired application. While significant research has been reported in the field of PDMS microfluidics, functionalization of PDMS surfaces remains a critical step in the fabrication process that is difficult to navigate. This review first offers a thorough illustration of existing fabrication methods for PDMS-based microfluidic devices, providing several recent advancements in this field with the aim of reducing the cost and time for mass production of these devices. Next, various conventional and emerging approaches for engineering the surface chemistry of PDMS are discussed in detail. We provide a wide range of functionalization techniques rendering PDMS microchannels highly biocompatible for physical or covalent immobilization of various biological entities while preventing non-specific interactions.
Collapse
Affiliation(s)
- Amid Shakeri
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada.
| | - Shadman Khan
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Tohid F Didar
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada.
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
26
|
Karan P, Chakraborty J, Chakraborty S, Wereley ST, Christov IC. Profiling a soft solid layer to passively control the conduit shape in a compliant microchannel during flow. Phys Rev E 2021; 104:015108. [PMID: 34412219 DOI: 10.1103/physreve.104.015108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 06/30/2021] [Indexed: 12/12/2022]
Abstract
The shape of a microchannel during flow through it is instrumental to understanding the physics that govern various phenomena ranging from rheological measurements of fluids to separation of particles and cells. Two commonly used approaches for obtaining a desired channel shape (for a given application) are (i) fabricating the microchannel in the requisite shape and (ii) actuating the microchannel walls during flow to obtain the requisite shape. However, these approaches are not always viable. We propose an alternative, passive approach to a priori tune the elastohydrodynamics in a microsystem toward achieving a predetermined (but not prefabricated) flow geometry when the microchannel is subjected to flow. That is, we use the interaction between a soft solid layer, the viscous flow beneath it, and the shaped rigid wall above it to tune the fluid domain's shape. Specifically, we study a parallel-wall microchannel whose top wall is a slender soft coating of arbitrary thickness attached to a rigid platform. We derive a nonlinear differential equation for the soft coating's fluid-solid interface, which we use to infer how to achieve specific conduit shapes during flow. Using this theory, we demonstrate the tuning of four categories of microchannel geometries, which establishes, via a proof-of-concept, the viability of our modeling framework. We also explore slip length patterning on the rigid bottom wall of the microchannel, a common technique in microfluidics, as an additional "handle" for microchannel shape control. However, we show that this effect is much weaker in practice.
Collapse
Affiliation(s)
- Pratyaksh Karan
- Department of Mechanical Engineering, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Jeevanjyoti Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Steven T Wereley
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Ivan C Christov
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| |
Collapse
|
27
|
Samiei M, Fathi M, Barar J, Fathi N, Amiryaghoubi N, Omidi Y. Bioactive hydrogel-based scaffolds for the regeneration of dental pulp tissue. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Pantermehl S, Emmert S, Foth A, Grabow N, Alkildani S, Bader R, Barbeck M, Jung O. 3D Printing for Soft Tissue Regeneration and Applications in Medicine. Biomedicines 2021; 9:biomedicines9040336. [PMID: 33810541 PMCID: PMC8066192 DOI: 10.3390/biomedicines9040336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
The use of additive manufacturing (AM) technologies is a relatively young research area in modern medicine. This technology offers a fast and effective way of producing implants, tissues, or entire organs individually adapted to the needs of a patient. Today, a large number of different 3D printing technologies with individual application areas are available. This review is intended to provide a general overview of these various printing technologies and their function for medical use. For this purpose, the design and functionality of the different applications are presented and their individual strengths and weaknesses are explained. Where possible, previous studies using the respective technologies in the field of tissue engineering are briefly summarized.
Collapse
Affiliation(s)
- Sven Pantermehl
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany; (S.P.); (S.E.); (A.F.); (O.J.)
| | - Steffen Emmert
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany; (S.P.); (S.E.); (A.F.); (O.J.)
| | - Aenne Foth
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany; (S.P.); (S.E.); (A.F.); (O.J.)
| | - Niels Grabow
- Institute for Biomedical Engineering, University Medical Center Rostock, 18119 Rostock, Germany;
| | | | - Rainer Bader
- Clinic and Policlinic for Orthopedics, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Mike Barbeck
- BerlinAnalytix GmbH, 12109 Berlin, Germany;
- Department of Ceramic Materials, Chair of Advanced Ceramic Materials, Institute for Materials Science and Technologies, Technical University Berlin, 10623 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)-17681022467
| | - Ole Jung
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany; (S.P.); (S.E.); (A.F.); (O.J.)
| |
Collapse
|
29
|
Sordi MB, Cruz A, Fredel MC, Magini R, Sharpe PT. Three-dimensional bioactive hydrogel-based scaffolds for bone regeneration in implant dentistry. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112055. [PMID: 33947549 DOI: 10.1016/j.msec.2021.112055] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022]
Abstract
Bone tissue requires a range of complex mechanisms to allow the restoration of its structure and function. Bone healing is a signaling cascade process, involving cells secreting cytokines, growth factors, and pro-inflammatory factors in the defect site that will, subsequently, recruit surrounding stem cells to migrate, proliferate, and differentiate into bone-forming cells. Bioactive functional scaffolds could be applied to improve the bone healing processes where the organism is not able to fully regenerate the lost tissue. However, to be optimal, such scaffolds should act as osteoconductors - supporting bone-forming cells, providing nutrients, and sustaining the arrival of new blood vessels, and act as osteoinducers - slowly releasing signaling molecules that stimulate mesenchymal stem cells to differentiate and deposit mineralized bone matrix. Different compositions and shapes of scaffolds, cutting-edge technologies, application of signaling molecules to promote cell differentiation, and high-quality biomaterials are reaching favorable outcomes towards osteoblastic differentiation of stem cells in in vitro and in vivo researches for bone regeneration. Hydrogel-based biomaterials are being pointed as promising for bone tissue regeneration; however, despite all the research and high-impact scientific publications, there are still several challenges that prevent the use of hydrogel-based scaffolds for bone regeneration being feasible for their clinical application. Hence, the objective of this review is to consolidate and report, based on the current scientific literature, the approaches for bone tissue regeneration using bioactive hydrogel-based scaffolds, cell-based therapies, and three-dimensional bioprinting to define the key challenges preventing their use in clinical applications.
Collapse
Affiliation(s)
- Mariane B Sordi
- Research Center on Dental Implants, Department of Odontology, Federal University of Santa Catarina, 88040-900 Florianopolis, SC, Brazil; Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, SE1 9RT, UK.
| | - Ariadne Cruz
- Department of Odontology, Federal University of Santa Catarina, 88040-900 Florianopolis, SC, Brazil.
| | - Márcio C Fredel
- Ceramic and Composite Materials Research Group, Department of Mechanical Engineering, Federal University of Santa Catarina, 88040-900 Florianopolis, SC, Brazil.
| | - Ricardo Magini
- Department of Odontology, Federal University of Santa Catarina, 88040-900 Florianopolis, SC, Brazil
| | - Paul T Sharpe
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, SE1 9RT, UK.
| |
Collapse
|
30
|
Sun M, Liu A, Yang X, Gong J, Yu M, Yao X, Wang H, He Y. 3D Cell Culture—Can It Be As Popular as 2D Cell Culture? ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Miao Sun
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - An Liu
- Department of Orthopaedic Surgery Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou 310000 China
| | - Xiaofu Yang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Jiaxing Gong
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Xinhua Yao
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology School of Stomatology Zhejiang University School of Medicine and Key Laboratory of Oral Biomedical Research of Zhejiang Province Hangzhou Zhejiang 310000 China
| | - Yong He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
- State Key Laboratory of Fluid Power and Mechatronic Systems School of Mechanical Engineering Zhejiang University Hangzhou 310000 China
| |
Collapse
|
31
|
Roshandel M, Dorkoosh F. Cardiac tissue engineering, biomaterial scaffolds, and their fabrication techniques. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5273] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Marjan Roshandel
- School of Chemical Engineering, College of Engineering University of Tehran Tehran Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy Tehran University of Medical Sciences Tehran Iran
- Medical Biomaterial Research Centre (MBRC) Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
32
|
Saska S, Pilatti L, Blay A, Shibli JA. Bioresorbable Polymers: Advanced Materials and 4D Printing for Tissue Engineering. Polymers (Basel) 2021; 13:563. [PMID: 33668617 PMCID: PMC7918883 DOI: 10.3390/polym13040563] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 01/10/2023] Open
Abstract
Three-dimensional (3D) printing is a valuable tool in the production of complexes structures with specific shapes for tissue engineering. Differently from native tissues, the printed structures are static and do not transform their shape in response to different environment changes. Stimuli-responsive biocompatible materials have emerged in the biomedical field due to the ability of responding to other stimuli (physical, chemical, and/or biological), resulting in microstructures modifications. Four-dimensional (4D) printing arises as a new technology that implements dynamic improvements in printed structures using smart materials (stimuli-responsive materials) and/or cells. These dynamic scaffolds enable engineered tissues to undergo morphological changes in a pre-planned way. Stimuli-responsive polymeric hydrogels are the most promising material for 4D bio-fabrication because they produce a biocompatible and bioresorbable 3D shape environment similar to the extracellular matrix and allow deposition of cells on the scaffold surface as well as in the inside. Subsequently, this review presents different bioresorbable advanced polymers and discusses its use in 4D printing for tissue engineering applications.
Collapse
Affiliation(s)
- Sybele Saska
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
| | - Livia Pilatti
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
| | - Alberto Blay
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
| | - Jamil Awad Shibli
- M3 Health Industria e Comercio de Produtos Medicos, Odontologicos e Correlatos S.A., Jundiaí, Sao Paulo 13212-213, Brazil; (S.S.); (L.P.); (A.B.)
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos, Sao Paulo 07023-070, Brazil
| |
Collapse
|
33
|
Sadeghianmaryan A, Naghieh S, Alizadeh Sardroud H, Yazdanpanah Z, Afzal Soltani Y, Sernaglia J, Chen X. Extrusion-based printing of chitosan scaffolds and their in vitro characterization for cartilage tissue engineering. Int J Biol Macromol 2020; 164:3179-3192. [DOI: 10.1016/j.ijbiomac.2020.08.180] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/18/2020] [Accepted: 08/22/2020] [Indexed: 01/01/2023]
|
34
|
Nie J, Fu J, He Y. Hydrogels: The Next Generation Body Materials for Microfluidic Chips? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003797. [PMID: 33103353 DOI: 10.1002/smll.202003797] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Indexed: 05/27/2023]
Abstract
The integration of microfluidics with biomedical research is confronted with considerable limitations due to its body materials. With high content of water, hydrogels own superior biocompatibility and degradability. Can hydrogels become another material choice for the construction of microfluidic chips, particularly biofluidics? The present review aims to systematically establish the concept of hydrogel-based microfluidic chips (HMCs) and address three main concerns: i) why choosing hydrogels? ii) how to fabricate HMCs?, and iii) in which fields to apply HMCs? It is envisioned that hydrogels may be used increasingly as substitute for traditional materials and gradually act as the body material for microfluidic chips. The modifications of conventional process are highlighted to overcome issues arising from the incompatibility between the construction methods and hydrogel materials. Specifically targeting at the "soft and wet" hydrogels, an efficient flowchart of "i) high resolution template printing; ii) damage-free demolding; iii) twice-crosslinking bonding" is proposed. Accordingly, a broader microfluidic chip concept is proposed in terms of form and function. Potential biomedical applications of HMCs are discussed. This review also highlights the challenges arising from the material replacement, as well as the future directions of the proposed concept. Finally, the authors' viewpoints and perspectives for this emerging field are discussed.
Collapse
Affiliation(s)
- Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
| |
Collapse
|
35
|
Fitzsimmons REB, Ireland RG, Zhong A, Soos A, Simmons CA. Assessment of fibrin-collagen co-gels for generating microvessels ex vivousing endothelial cell-lined microfluidics and multipotent stromal cell (MSC)-induced capillary morphogenesis. Biomed Mater 2020; 16. [PMID: 33086195 DOI: 10.1088/1748-605x/abc38f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/21/2020] [Indexed: 01/28/2023]
Abstract
One aspect of the challenge of engineering viable tissues ex vivo is the generation of perfusable microvessels of varying diameters. In this work, we take the approach of using hydrogel-based microfluidics seeded with endothelial cells (ECs) to form small artery/vein-like vessels, in conjunction with using the self-assembly behavior of ECs to form capillary-like vessels when co-cultured with multipotent stromal cells (MSCs). In exploring this approach, we focused on investigating collagen, fibrin, and various collagen-fibrin co-gel formulations for their potential suitability as serving as scaffold materials by surveying their angiogencity and mechanical properties. Fibrin and co-gels successfully facilitated multicellular EC sprouting, whereas collagen elicited a migration response of individual ECs, unless supplemented with the PKC (protein kinase C)-activator, phorbol 12-myristate 13-acetate. Collagen scaffolds were also found to severely contract when embedded with mesenchymal cells, but this contraction could be abrogated with the addition of fibrin. Increasing collagen content within co-gel formulations, however, imparted a higher compressive modulus and allowed for the reliable formation of intact hydrogel-based microchannels which could then be perfused. Given the bioactivity and mechanical benefits of fibrin and collagen, respectively, collagen-fibrin co-gels are a promising scaffold option for generating vascularized tissue constructs.
Collapse
Affiliation(s)
- Ross E B Fitzsimmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, CANADA
| | - Ronald G Ireland
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, CANADA
| | - Aileen Zhong
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, CANADA
| | - Agnes Soos
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, CANADA
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, CANADA
| |
Collapse
|
36
|
Zhang W, Huang G, Xu F. Engineering Biomaterials and Approaches for Mechanical Stretching of Cells in Three Dimensions. Front Bioeng Biotechnol 2020; 8:589590. [PMID: 33154967 PMCID: PMC7591716 DOI: 10.3389/fbioe.2020.589590] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Mechanical stretch is widely experienced by cells of different tissues in the human body and plays critical roles in regulating their behaviors. Numerous studies have been devoted to investigating the responses of cells to mechanical stretch, providing us with fruitful findings. However, these findings have been mostly observed from two-dimensional studies and increasing evidence suggests that cells in three dimensions may behave more closely to their in vivo behaviors. While significant efforts and progresses have been made in the engineering of biomaterials and approaches for mechanical stretching of cells in three dimensions, much work remains to be done. Here, we briefly review the state-of-the-art researches in this area, with focus on discussing biomaterial considerations and stretching approaches. We envision that with the development of advanced biomaterials, actuators and microengineering technologies, more versatile and predictive three-dimensional cell stretching models would be available soon for extensive applications in such fields as mechanobiology, tissue engineering, and drug screening.
Collapse
Affiliation(s)
- Weiwei Zhang
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Guoyou Huang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing University, Chongqing, China
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center, Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
37
|
Correa SO, Luo X, Raub CB. Microfluidic fabrication of stable collagen microgels with aligned microstructure using flow-driven co-deposition and ionic gelation. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2020; 30:085002. [PMID: 37273664 PMCID: PMC10237176 DOI: 10.1088/1361-6439/ab8ebf] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The controlled biofabrication of stable, aligned collagen hydrogels within microfluidic devices is critically important to the design of more physiologically accurate, longer-cultured on-chip models of tissue and organs. To address this goal, collagen-alginate microgels were formed in a microfluidic channel by calcium crosslinking of a flowing collagen-alginate solution through a cross-channel chitosan membrane spanning a pore allowing ion diffusion but not convection. The gels formed within seconds as isolated islands in a single channel, and their growth was self-limiting. Total gel thickness was controlled by altering the concentration of calcium and collagen-alginate flow rate to reach an equilibrium of calcium diffusion and solution convection at the gel boundary, for a desired thickness of 30-200 μm. Additionally, less calcium and higher flow produced greater compression of the gel, with regions farther from the pore compressing more. An aligned, stable collagen network was demonstrated by collagen birefringence, circumferential texture orientation, and little change in gel dimensions with de-chelation of calcium from alginate by prolonged flow of EDTA in the channel. Resultant gels were most stable and only slightly asymmetric when formed from solutions containing 8 mg ml-1 collagen. Diffusion of 4 kDa and 70 kDa fluorescently-labeled dextran indicated size-dependent diffusion across the gel, and accessibility of the construct to appropriately-sized bioactive molecules. This work demonstrates the physicochemical parameter control of collagen gel formation in microfluidic devices, with utility toward on-chip models of dense extracellular matrix invasion, cancer growth and drug delivery to cells within dense extracellular matrix bodies.
Collapse
Affiliation(s)
- Santiago O Correa
- Department of Biomedical Engineering, Washington DC, United States of America
| | - Xiaolong Luo
- Department of Mechanical Engineering, Washington DC, United States of America
- These authors contributed equally to this work
| | - Christopher B Raub
- Department of Biomedical Engineering, Washington DC, United States of America
- These authors contributed equally to this work
| |
Collapse
|
38
|
Shen C, Li Y, Wang Y, Meng Q. Non-swelling hydrogel-based microfluidic chips. LAB ON A CHIP 2019; 19:3962-3973. [PMID: 31656966 DOI: 10.1039/c9lc00564a] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Hydrogel-based microfluidic chips are more biologically relevant than conventional polydimethylsiloxane (PDMS) chips, but the inherent swelling of hydrogels leads to a decrease in mechanical performance and deformation of the as-prepared structure in their manufacture and application processing. Non-swelling hydrogel has, for the first time, been utilized to construct microfluidic chips in this study. It was fabricated by covalently cross-linking the biocompatible copolymer of di-acrylated Pluronic F127 (F127-DA). Thanks to their non-swelling property, the hydrogel-based microfluidic chips maintain their as-prepared mechanical strength and channel morphology when equilibrated in aqueous solution at 37 °C. Moreover, the microfluidic chips are autoclavable and show an appropriately slow degradation rate by remaining stable within 21 days of incubation. Based on these properties, a vessel-on-a-chip was established by seeding human umbilical vein endothelial cells (HUVECs) onto the microchannel surfaces inside the microfluidic chip. Under 6 days of perfusion culture with a physiologically relevant shear stress of 5 dyne per cm2, the HUVECs in the chip show responsivity to fluid shear stress and express higher endothelial functions than the corresponding static culture. Therefore, non-swelling hydrogel-based microfluidic chips could potentially be applicable for cell/tissue-related applications, performing much better than conventional PDMS or existing hydrogel based microfluidic chips.
Collapse
Affiliation(s)
- Chong Shen
- College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, PR China.
| | - Yingjun Li
- College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, PR China.
| | - Ying Wang
- College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, PR China.
| | - Qin Meng
- College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, PR China.
| |
Collapse
|
39
|
O’Grady BJ, Balikov DA, Lippmann ES, Bellan LM. Spatiotemporal Control of Morphogen Delivery to Pattern Stem Cell Differentiation in Three-Dimensional Hydrogels. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2019; 51:e97. [PMID: 31756050 PMCID: PMC6876696 DOI: 10.1002/cpsc.97] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Morphogens are biological molecules that alter cellular identity and behavior across both space and time. During embryonic development, morphogen spatial localization can be confined to small volumes in a single tissue or permeate throughout an entire organism, and the temporal effects of morphogens can range from fractions of a second to several days. In most cases, morphogens are presented as a gradient to adjacent cells within tissues to pattern cell fate. As such, to appropriately model development and build representative multicellular architectures in vitro, it is vital to recapitulate these gradients during stem cell differentiation. However, the ability to control morphogen presentation within in vitro systems remains challenging. Here, we describe an innovative platform using channels patterned within thick, three-dimensional hydrogels that deliver multiple morphogens to embedded cells, thereby demonstrating exquisite control over both spatial and temporal variations in morphogen presentation. This generalizable approach should have broad utility for researchers interested in patterning in vitro tissue structures. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Brian J. O’Grady
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
- BJO and DAB contributed equally to this work as co-first authors
| | - Daniel A. Balikov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- BJO and DAB contributed equally to this work as co-first authors
| | - Ethan S. Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- ESL and LMB contributed equally to this work as co-senior authors
| | - Leon M. Bellan
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- ESL and LMB contributed equally to this work as co-senior authors
| |
Collapse
|
40
|
Mantha S, Pillai S, Khayambashi P, Upadhyay A, Zhang Y, Tao O, Pham HM, Tran SD. Smart Hydrogels in Tissue Engineering and Regenerative Medicine. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E3323. [PMID: 31614735 PMCID: PMC6829293 DOI: 10.3390/ma12203323] [Citation(s) in RCA: 412] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 01/01/2023]
Abstract
The field of regenerative medicine has tremendous potential for improved treatment outcomes and has been stimulated by advances made in bioengineering over the last few decades. The strategies of engineering tissues and assembling functional constructs that are capable of restoring, retaining, and revitalizing lost tissues and organs have impacted the whole spectrum of medicine and health care. Techniques to combine biomimetic materials, cells, and bioactive molecules play a decisive role in promoting the regeneration of damaged tissues or as therapeutic systems. Hydrogels have been used as one of the most common tissue engineering scaffolds over the past two decades due to their ability to maintain a distinct 3D structure, to provide mechanical support for the cells in the engineered tissues, and to simulate the native extracellular matrix. The high water content of hydrogels can provide an ideal environment for cell survival, and structure which mimics the native tissues. Hydrogel systems have been serving as a supportive matrix for cell immobilization and growth factor delivery. This review outlines a brief description of the properties, structure, synthesis and fabrication methods, applications, and future perspectives of smart hydrogels in tissue engineering.
Collapse
Affiliation(s)
- Somasundar Mantha
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Sangeeth Pillai
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Parisa Khayambashi
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Akshaya Upadhyay
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Yuli Zhang
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Owen Tao
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Hieu M Pham
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, 3640 University Street, Montreal, QC H3A 0C7, Canada.
| |
Collapse
|
41
|
Fustin JM, Li M, Gao B, Chen Q, Cheng T, Stewart AG. Rhythm on a chip: circadian entrainment in vitro is the next frontier in body-on-a chip technology. Curr Opin Pharmacol 2019; 48:127-136. [PMID: 31600661 DOI: 10.1016/j.coph.2019.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 01/01/2023]
Abstract
Organoids, bioprinted mini-tissues and body-on-a-chip technologies are poised to transform the practice of preclinical pharmacology, with a view to achieving better predictive value. We review the need for further refinement in static and dynamic biomechanical aspects of such microenvironments. Further consideration of the developments required in perfusion systems to enable delivery of an appropriate soluble microenvironment are argued. We place particular emphasis on a major deficiency in these systems, being the absence or aberrant circadian behaviour of cells used in such settings, and consider the technical challenges that are needing to be met in order to achieve rhythm-on-a-chip.
Collapse
Affiliation(s)
- Jean-Michel Fustin
- Laboratory of Molecular Metabology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Meina Li
- ARC Centre for Personalised Therapeutics Technologies, Department of Pharmacology & Therapeutics, School of Biomedical Science, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Bryan Gao
- ARC Centre for Personalised Therapeutics Technologies, Department of Pharmacology & Therapeutics, School of Biomedical Science, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Qianyu Chen
- ARC Centre for Personalised Therapeutics Technologies, Department of Pharmacology & Therapeutics, School of Biomedical Science, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tianhong Cheng
- ARC Centre for Personalised Therapeutics Technologies, Department of Pharmacology & Therapeutics, School of Biomedical Science, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alastair G Stewart
- ARC Centre for Personalised Therapeutics Technologies, Department of Pharmacology & Therapeutics, School of Biomedical Science, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
42
|
Abstract
Biofabrication techniques have enabled the formation of complex models of many biological tissues. We present a framework to contextualize biofabrication techniques within a disease modeling application. Fibrosis is a progressive disease interfering with tissue structure and function, which stems from an aberrant wound healing response. Epithelial injury and clot formation lead to fibroblast invasion and activation, followed by contraction and remodeling of the extracellular matrix. These stages have healthy wound healing variants in addition to the pathogenic analogs that are seen in fibrosis. This review evaluates biofabrication of a variety of phenotypic cell-based fibrosis assays. By recapitulating different contributors to fibrosis, these assays are able to evaluate biochemical pathways and therapeutic candidates for specific stages of fibrosis pathogenesis. Biofabrication of these culture models may enable phenotypic screening for improved understanding of fibrosis biology as well as improved screening of anti-fibrotic therapeutics.
Collapse
Affiliation(s)
- Cameron Yamanishi
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, United States of America
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, United States of America
| | - Stephen Robinson
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, United States of America
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, United States of America
| | - Shuichi Takayama
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, United States of America
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, United States of America
| |
Collapse
|
43
|
Taira N, Ino K, Ida H, Nashimoto Y, Shiku H. Electrodeposition-based rapid bioprinting of 3D-designed hydrogels with a pin art device. Biofabrication 2019; 11:035018. [DOI: 10.1088/1758-5090/ab166e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
44
|
Indirect 3D bioprinting and characterization of alginate scaffolds for potential nerve tissue engineering applications. J Mech Behav Biomed Mater 2019; 93:183-193. [DOI: 10.1016/j.jmbbm.2019.02.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 11/05/2018] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
|
45
|
Chen S, Zhao Y, Yan X, Zhang L, Li G, Yang Y. PAM/GO/gel/SA composite hydrogel conduit with bioactivity for repairing peripheral nerve injury. J Biomed Mater Res A 2019; 107:1273-1283. [DOI: 10.1002/jbm.a.36637] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/02/2019] [Accepted: 01/28/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Shiyu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong University 226001, Nantong People's Republic of China
- Co‐innovation Center of NeuroregenerationNantong University 226001, Nantong People's Republic of China
| | - Yinxin Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong University 226001, Nantong People's Republic of China
- Co‐innovation Center of NeuroregenerationNantong University 226001, Nantong People's Republic of China
| | - Xiaoli Yan
- Jiangsu Testing and Inspection Institute for Medical Devices 17 Kangwen Road, Nanjing JS 210019 People's Republic of China
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong University 226001, Nantong People's Republic of China
- Co‐innovation Center of NeuroregenerationNantong University 226001, Nantong People's Republic of China
| | - Guicai Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong University 226001, Nantong People's Republic of China
- Co‐innovation Center of NeuroregenerationNantong University 226001, Nantong People's Republic of China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong University 226001, Nantong People's Republic of China
- Co‐innovation Center of NeuroregenerationNantong University 226001, Nantong People's Republic of China
| |
Collapse
|
46
|
Witzel II, Nasser R, Garcia-Sabaté A, Sapudom J, Ma C, Chen W, Teo JCM. Deconstructing Immune Microenvironments of Lymphoid Tissues for Reverse Engineering. Adv Healthc Mater 2019; 8:e1801126. [PMID: 30516005 DOI: 10.1002/adhm.201801126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Indexed: 01/01/2023]
Abstract
The immune microenvironment presents a diverse panel of cues that impacts immune cell migration, organization, differentiation, and the immune response. Uniquely, both the liquid and solid phases of every specific immune niche within the body play an important role in defining cellular functions in immunity at that particular location. The in vivo immune microenvironment consists of biomechanical and biochemical signals including their gradients, surface topography, dimensionality, modes of ligand presentation, and cell-cell interactions, and the ability to recreate these immune biointerfaces in vitro can provide valuable insights into the immune system. This manuscript reviews the critical roles played by different immune cells and surveys the current progress of model systems for reverse engineering of immune microenvironments with a focus on lymphoid tissues.
Collapse
Affiliation(s)
- Ini-Isabée Witzel
- Core Technology Platforms; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Rasha Nasser
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
- Department of Biomedical Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| | - Jeremy C. M. Teo
- Laboratory for Immuno Bioengineering Research and Applications (LIBRA); Division of Engineering; New York University Abu Dhabi; Saadiyat Campus, P.O. Box 127788 Abu Dhabi UAE
- Department of Mechanical and Aerospace Engineering; New York University; 6 MetroTech Center Brooklyn NY 11201 USA
| |
Collapse
|
47
|
Christoffersson J, Aronsson C, Jury M, Selegård R, Aili D, Mandenius CF. Fabrication of modular hyaluronan-PEG hydrogels to support 3D cultures of hepatocytes in a perfused liver-on-a-chip device. Biofabrication 2018; 11:015013. [DOI: 10.1088/1758-5090/aaf657] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
48
|
Akay M, Hite J, Avci NG, Fan Y, Akay Y, Lu G, Zhu JJ. Drug Screening of Human GBM Spheroids in Brain Cancer Chip. Sci Rep 2018; 8:15423. [PMID: 30337660 PMCID: PMC6194126 DOI: 10.1038/s41598-018-33641-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), an extremely invasive and high-grade (grade IV) glioma, is the most common and aggressive form of brain cancer. It has a poor prognosis, with a median overall survival of only 11 months in the general GBM population and 14.6 to 21 months in clinical trial participants with standard GBM therapies, including maximum safe craniotomy, adjuvant radiation, and chemotherapies. Therefore, new approaches for developing effective treatments, such as a tool for assessing tumor cell drug response before drug treatments are administered, are urgently needed to improve patient survival. To address this issue, we developed an improved brain cancer chip with a diffusion prevention mechanism that blocks drugs crossing from one channel to another. In the current study, we demonstrate that the chip has the ability to culture 3D spheroids from patient tumor specimen-derived GBM cells obtained from three GBM patients. Two clinical drugs used to treat GBM, temozolomide (TMZ) and bevacizumab (Avastin, BEV), were applied and a range of relative concentrations was generated by the microfluidic channels in the brain cancer chip. The results showed that TMZ works more effectively when used in combination with BEV compared to TMZ alone. We believe that this low-cost brain cancer chip could be further developed to generate optimal combination of chemotherapy drugs tailored to individual GBM patients.
Collapse
Affiliation(s)
- Metin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA.
| | - John Hite
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Naze Gul Avci
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yantao Fan
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Yasemin Akay
- Department of Biomedical Engineering, University of Houston, 3517 Cullen Blvd, Houston, TX, USA
| | - Guangrong Lu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| | - Jay-Jiguang Zhu
- Mischer Neuroscience Associates and the Vivian L. Smith Department of Neurosurgery University of Texas Health Science Center in Houston, UTHealth and Memorial Hermann, 6400 Fannin St. Suite 2800, Houston, TX, 77030, USA
| |
Collapse
|
49
|
Genderen AM, Jansen J, Cheng C, Vermonden T, Masereeuw R. Renal Tubular- and Vascular Basement Membranes and their Mimicry in Engineering Vascularized Kidney Tubules. Adv Healthc Mater 2018; 7:e1800529. [PMID: 30091856 DOI: 10.1002/adhm.201800529] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/18/2018] [Indexed: 01/09/2023]
Abstract
The high prevalence of chronic kidney disease leads to an increased need for renal replacement therapies. While there are simply not enough donor organs available for transplantation, there is a need to seek other therapeutic avenues as current dialysis modalities are insufficient. The field of regenerative medicine and whole organ engineering is emerging, and researchers are looking for innovative ways to create (part of) a functional new organ. To biofabricate a kidney or its functional units, it is necessary to understand and learn from physiology to be able to mimic the specific tissue properties. Herein is provided an overview of the knowledge on tubular and vascular basement membranes' biochemical components and biophysical properties, and the major differences between the two basement membranes are highlighted. Furthermore, an overview of current trends in membrane technology for developing renal replacement therapies and to stimulate kidney regeneration is provided.
Collapse
Affiliation(s)
- Anne Metje Genderen
- Division of PharmacologyUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| | - Jitske Jansen
- Division of PharmacologyUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| | - Caroline Cheng
- Regenerative Medicine Center UtrechtUniversity Medical Center Utrecht 3584 CT Utrecht The Netherlands
- Department of Nephrology and HypertensionUniversity Medical Center Utrecht 3508 GA Utrecht The Netherlands
- Department of Experimental CardiologyErasmus Medical Center 3015 GD Rotterdam The Netherlands
| | - Tina Vermonden
- Division of PharmaceuticsUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| | - Rosalinde Masereeuw
- Division of PharmacologyUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| |
Collapse
|
50
|
Abstract
Vascularization is essential for tissue regeneration. Despite extensive efforts in the past decades, sufficient and rapid vascularization remains a major challenge in tissue engineering. Many studies have shown that the addition of channels in a porous scaffold can provide the ability to promote cell growth and rapid vascularization, thus leading to better outcomes in new tissue formation. Large size scaffolds lack perfusable channel networks and negatively impair the survival of transplanted cells and tissue function development, leading to necrotic core formation and the failure of functional tissue formation. Presently, there are many methods to produce channels in porous scaffolds for vascularization. Here, we review the function of channels in porous scaffolds and the approaches to produce those channels.
Collapse
Affiliation(s)
- Yunqing Kang
- Department of Ocean & Mechanical Engineering, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA.,Department of Biomedical Science, College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Jia Chang
- Department of Periodontology, University of Florida College of Dentistry, Gainesville, FL 32610, USA
| |
Collapse
|