1
|
Rashed MS, Abdelkarim EA, Elsamahy T, Sobhy M, El-Mesery HS, Salem A. Advances in cell-based biosensors: Transforming food flavor evaluation with novel approaches. Food Chem X 2025; 26:102336. [PMID: 40115496 PMCID: PMC11923814 DOI: 10.1016/j.fochx.2025.102336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025] Open
Abstract
Food flavor, a blend of taste and smell, is key to consumer acceptance and food quality. Traditional sensory and instrumental methods often fail to replicate human sensory responses. This review discusses the role of cell-based biosensors in flavor evaluation, showcasing their sensitivity, specificity, and rapid response. Using living cells like taste and olfactory cells, these biosensors surpass traditional approaches. Advancements include microelectrode array systems with taste receptor cells for real-time detection of bitter, sweet, and umami substances and improved cell immobilization technologies for detecting complex odorant profiles. Challenges such as signal stability, selective detection, cell cultivation, and scalability persist. However, integrating artificial intelligence and portable technologies could broaden their applications. With the potential to revolutionize sensory analysis, cell-based biosensors offer a sustainable, precise, and scalable approach to food flavor evaluation, bridging sensory perception with advanced analytical methods and driving innovation in food science.
Collapse
Affiliation(s)
- Mahmoud Said Rashed
- Food Science and Technology Department, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
| | - Esraa A Abdelkarim
- Food Control Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | | | - Mabrouk Sobhy
- Food Science and Technology Department, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
- School of Agricultural Engineering, Jiangsu University, Zhenjiang 212013, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Hany S El-Mesery
- School of Energy and Power Engineering, Jiangsu University, Zhenjiang 212013, China
- Agricultural Engineering Research Institute, Agricultural Research Center, Dokki, 12611 Giza, Egypt
| | - Ali Salem
- Civil Engineering Department, Faculty of Engineering, Minia University, Minia 61111, Egypt
- Structural Diagnostics and Analysis Research Group, Faculty of Engineering and Information Technology, University of Pecs, Hungary
| |
Collapse
|
2
|
Nejati B, Shahhosseini R, Hajiabbasi M, Ardabili NS, Baktash KB, Alivirdiloo V, Moradi S, Rad MF, Rahimi F, Farani MR, Ghazi F, Mobed A, Alipourfard I. Cancer-on-chip: a breakthrough organ-on-a-chip technology in cancer cell modeling. Med Biol Eng Comput 2025; 63:321-337. [PMID: 39400856 PMCID: PMC11750902 DOI: 10.1007/s11517-024-03199-5] [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: 01/12/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024]
Abstract
Cancer remains one of the leading causes of death worldwide. The unclear molecular mechanisms and complex in vivo microenvironment of tumors make it difficult to clarify the nature of cancer and develop effective treatments. Therefore, the development of new methods to effectively treat cancer is urgently needed and of great importance. Organ-on-a-chip (OoC) systems could be the breakthrough technology sought by the pharmaceutical industry to address ever-increasing research and development costs. The past decade has seen significant advances in the spatial modeling of cancer therapeutics related to OoC technology, improving physiological exposition criteria. This article aims to summarize the latest achievements and research results of cancer cell treatment simulated in a 3D microenvironment using OoC technology. To this end, we will first discuss the OoC system in detail and then demonstrate the latest findings of the cancer cell treatment study by Ooc and how this technique can potentially optimize better modeling of the tumor. The prospects of OoC systems in the treatment of cancer cells and their advantages and limitations are also among the other points discussed in this study.
Collapse
Affiliation(s)
- Babak Nejati
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | - Vahid Alivirdiloo
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Sadegh Moradi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Fatemeh Rahimi
- Division of Clinical Laboratory, Zahra Mardani Azar Children Training Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhood Ghazi
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mobed
- Department of Community Medicine, Faculty of Medicine, Social Determinants of Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
3
|
Micati D, Hlavca S, Chan WH, Abud HE. Harnessing 3D models to uncover the mechanisms driving infectious and inflammatory disease in the intestine. BMC Biol 2024; 22:300. [PMID: 39736603 DOI: 10.1186/s12915-024-02092-9] [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: 06/16/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
Representative models of intestinal diseases are transforming our knowledge of the molecular mechanisms of disease, facilitating effective drug screening and avenues for personalised medicine. Despite the emergence of 3D in vitro intestinal organoid culture systems that replicate the genetic and functional characteristics of the epithelial tissue of origin, there are still challenges in reproducing the human physiological tissue environment in a format that enables functional readouts. Here, we describe the latest platforms engineered to investigate environmental tissue impacts, host-microbe interactions and enable drug discovery. This highlights the potential to revolutionise knowledge on the impact of intestinal infection and inflammation and enable personalised disease modelling and clinical translation.
Collapse
Affiliation(s)
- Diana Micati
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Sara Hlavca
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Wing Hei Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
4
|
Moro LG, Guarnier LP, Azevedo MF, Fracasso JAR, Lucio MA, de Castro MV, Dias ML, Lívero FADR, Ribeiro-Paes JT. A Brief History of Cell Culture: From Harrison to Organs-on-a-Chip. Cells 2024; 13:2068. [PMID: 39768159 PMCID: PMC11674496 DOI: 10.3390/cells13242068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 01/11/2025] Open
Abstract
This comprehensive overview of the historical milestones in cell culture underscores key breakthroughs that have shaped the field over time. It begins with Wilhelm Roux's seminal experiments in the 1880s, followed by the pioneering efforts of Ross Granville Harrison, who initiated groundbreaking experiments that fundamentally shaped the landscape of cell culture in the early 20th century. Carrel's influential contributions, notably the immortalization of chicken heart cells, have marked a significant advancement in cell culture techniques. Subsequently, Johannes Holtfreter, Aron Moscona, and Joseph Leighton introduced methodological innovations in three-dimensional (3D) cell culture, initiated by Alexis Carrel, laying the groundwork for future consolidation and expansion of the use of 3D cell culture in different areas of biomedical sciences. The advent of induced pluripotent stem cells by Takahashi and Yamanaka in 2006 was revolutionary, enabling the reprogramming of differentiated cells into a pluripotent state. Since then, recent innovations have included spheroids, organoids, and organ-on-a-chip technologies, aiming to mimic the structure and function of tissues and organs in vitro, pushing the boundaries of biological modeling and disease understanding. In this review, we overview the history of cell culture shedding light on the main discoveries, pitfalls and hurdles that were overcome during the transition from 2D to 3D cell culture techniques. Finally, we discussed the future directions for cell culture research that may accelerate the development of more effective and personalized treatments.
Collapse
Affiliation(s)
- Lincoln Gozzi Moro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Lucas Pires Guarnier
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
| | | | | | - Marco Aurélio Lucio
- Graduate Program in Environment and Regional Development, University of Western São Paulo, Presidente Prudente 19050-920, Brazil;
| | - Mateus Vidigal de Castro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Marlon Lemos Dias
- Precision Medicine Research Center, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro—UFRJ, Rio de Janeiro 21941-630, Brazil;
| | | | - João Tadeu Ribeiro-Paes
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
- Laboratory of Genetics and Cell Therapy (GenTe Cel), Department of Biotechnology, São Paulo State University—UNESP, Assis 19806-900, Brazil
| |
Collapse
|
5
|
Urciuolo F, Imparato G, Netti PA. Engineering Cell Instructive Microenvironments for In Vitro Replication of Functional Barrier Organs. Adv Healthc Mater 2024; 13:e2400357. [PMID: 38695274 DOI: 10.1002/adhm.202400357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/02/2024] [Indexed: 05/14/2024]
Abstract
Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
| | - Giorgia Imparato
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| |
Collapse
|
6
|
Fritschen A, Lindner N, Scholpp S, Richthof P, Dietz J, Linke P, Guttenberg Z, Blaeser A. High-Scale 3D-Bioprinting Platform for the Automated Production of Vascularized Organs-on-a-Chip. Adv Healthc Mater 2024; 13:e2304028. [PMID: 38511587 PMCID: PMC11469029 DOI: 10.1002/adhm.202304028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/18/2024] [Indexed: 03/22/2024]
Abstract
3D bioprinting possesses the potential to revolutionize contemporary methodologies for fabricating tissue models employed in pharmaceutical research and experimental investigations. This is enhanced by combining bioprinting with advanced organs-on-a-chip (OOCs), which includes a complex arrangement of multiple cell types representing organ-specific cells, connective tissue, and vasculature. However, both OOCs and bioprinting so far demand a high degree of manual intervention, thereby impeding efficiency and inhibiting scalability to meet technological requirements. Through the combination of drop-on-demand bioprinting with robotic handling of microfluidic chips, a print procedure is achieved that is proficient in managing three distinct tissue models on a chip within only a minute, as well as capable of consecutively processing numerous OOCs without manual intervention. This process rests upon the development of a post-printing sealable microfluidic chip, that is compatible with different types of 3D-bioprinters and easily connected to a perfusion system. The capabilities of the automized bioprint process are showcased through the creation of a multicellular and vascularized liver carcinoma model on the chip. The process achieves full vascularization and stable microvascular network formation over 14 days of culture time, with pronounced spheroidal cell growth and albumin secretion of HepG2 serving as a representative cell model.
Collapse
Affiliation(s)
- Anna Fritschen
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Nils Lindner
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Sebastian Scholpp
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Philipp Richthof
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | - Jonas Dietz
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
| | | | | | - Andreas Blaeser
- BioMedical Printing TechnologyDepartment of Mechanical EngineeringTechnical University of Darmstadt64289DarmstadtGermany
- Centre for Synthetic BiologyTechnical University of Darmstadt64289DarmstadtGermany
| |
Collapse
|
7
|
De Spirito M, Palmieri V, Perini G, Papi M. Bridging the Gap: Integrating 3D Bioprinting and Microfluidics for Advanced Multi-Organ Models in Biomedical Research. Bioengineering (Basel) 2024; 11:664. [PMID: 39061746 PMCID: PMC11274229 DOI: 10.3390/bioengineering11070664] [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: 05/28/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Recent advancements in 3D bioprinting and microfluidic lab-on-chip systems offer promising solutions to the limitations of traditional animal models in biomedical research. Three-dimensional bioprinting enables the creation of complex, patient-specific tissue models that mimic human physiology more accurately than animal models. These 3D bioprinted tissues, when integrated with microfluidic systems, can replicate the dynamic environment of the human body, allowing for the development of multi-organ models. This integration facilitates more precise drug screening and personalized therapy development by simulating interactions between different organ systems. Such innovations not only improve predictive accuracy but also address ethical concerns associated with animal testing, aligning with the three Rs principle. Future directions include enhancing bioprinting resolution, developing advanced bioinks, and incorporating AI for optimized system design. These technologies hold the potential to revolutionize drug development, regenerative medicine, and disease modeling, leading to more effective, personalized, and humane treatments.
Collapse
Affiliation(s)
- Marco De Spirito
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Valentina Palmieri
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
- Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche, CNR, via dei Taurini 19, 00185 Rome, Italy
| | - Giordano Perini
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Massimiliano Papi
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
8
|
Vashishat A, Patel P, Das Gupta G, Das Kurmi B. Alternatives of Animal Models for Biomedical Research: a Comprehensive Review of Modern Approaches. Stem Cell Rev Rep 2024; 20:881-899. [PMID: 38429620 DOI: 10.1007/s12015-024-10701-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
Biomedical research has long relied on animal models to unravel the intricacies of human physiology and pathology. However, concerns surrounding ethics, expenses, and inherent species differences have catalyzed the exploration of alternative avenues. The contemporary alternatives to traditional animal models in biomedical research delve into three main categories of alternative approaches: in vitro models, in vertebrate models, and in silico models. This unique approach to artificial intelligence and machine learning has been a keen interest to be used in different biomedical research. The main goal of this review is to serve as a guide to researchers seeking novel avenues for their investigations and underscores the importance of considering alternative models in the pursuit of scientific knowledge and medical breakthroughs, including showcasing the broad spectrum of modern approaches that are revolutionizing biomedical research and leading the way toward a more ethical, efficient, and innovative future. Models can insight into cellular processes, developmental biology, drug interaction, assessing toxicology, and understanding molecular mechanisms.
Collapse
Affiliation(s)
- Abhinav Vashishat
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
9
|
Arumugam P, Kaarthikeyan G, Eswaramoorthy R. Three-Dimensional Bioprinting: The Ultimate Pinnacle of Tissue Engineering. Cureus 2024; 16:e58029. [PMID: 38738080 PMCID: PMC11088218 DOI: 10.7759/cureus.58029] [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: 02/27/2024] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Three-dimensional (3D) bioprinting has emerged as a revolutionary additive manufacturing technology that can potentially enable life-changing medical treatments in regenerative medicine. It applies the principles of tissue engineering for the printing of tissues and organs in a layer-by-layer manner. This review focuses on the various 3D bioprinting technologies currently available, the different biomaterials, cells, and growth factors that can be utilized to develop tissue-specific bioinks, the different venues for applying these technologies, and the challenges this technology faces.
Collapse
Affiliation(s)
- Parkavi Arumugam
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - G Kaarthikeyan
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Rajalakshmanan Eswaramoorthy
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
10
|
Yasli M, Dabbagh SR, Tasoglu S, Aydin S. Additive manufacturing and three-dimensional printing in obstetrics and gynecology: a comprehensive review. Arch Gynecol Obstet 2023; 308:1679-1690. [PMID: 36635490 DOI: 10.1007/s00404-023-06912-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023]
Abstract
Three-dimensional (3D) printing, also known as additive manufacturing, is a technology used to create complex 3D structures out of a digital model that can be almost any shape. Additive manufacturing allows the creation of customized, finely detailed constructs. Improvements in 3D printing, increased 3D printer availability, decreasing costs, development of biomaterials, and improved cell culture techniques have enabled complex, novel, and customized medical applications to develop. There have been rapid development and utilization of 3D printing technologies in orthopedics, dentistry, urology, reconstructive surgery, and other health care areas. Obstetrics and Gynecology (OBGYN) is an emerging application field for 3D printing. This technology can be utilized in OBGYN for preventive medicine, early diagnosis, and timely treatment of women-and-fetus-specific health issues. Moreover, 3D printed simulations of surgical procedures enable the training of physicians according to the needs of any given procedure. Herein, we summarize the technology and materials behind additive manufacturing and review the most recent advancements in the application of 3D printing in OBGYN studies, such as diagnosis, surgical planning, training, simulation, and customized prosthesis. Furthermore, we aim to give a future perspective on the integration of 3D printing and OBGYN applications and to provide insight into the potential applications.
Collapse
Affiliation(s)
- Mert Yasli
- Koç University School of Medicine, Koç University, Sariyer, 34450, Istanbul, Turkey
| | - Sajjad Rahmani Dabbagh
- Department of Mechanical Engineering, Koç University, Sariyer, 34450, Istanbul, Turkey
- Arçelik Research Center for Creative Industries (KUAR), Koç University, Koç University, Sariyer, 3445, Istanbul, Turkey
- Koc University Is Bank Artificial Intelligence Lab (KUIS AILab), Koç University, Sariyer, 34450, Istanbul, Turkey
| | - Savas Tasoglu
- Department of Mechanical Engineering, Koç University, Sariyer, 34450, Istanbul, Turkey
- Arçelik Research Center for Creative Industries (KUAR), Koç University, Koç University, Sariyer, 3445, Istanbul, Turkey
- Koc University Is Bank Artificial Intelligence Lab (KUIS AILab), Koç University, Sariyer, 34450, Istanbul, Turkey
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Serdar Aydin
- Department of Obstetrics and Gynecology, Koç University Hospital, Davutpaşa Cad. No:4, Zeytinburnu, 34010, Istanbul, Turkey.
- Koç University School of Medicine, Koç University, Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
11
|
Bakhshandeh B, Sorboni SG, Ranjbar N, Deyhimfar R, Abtahi MS, Izady M, Kazemi N, Noori A, Pennisi CP. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp Cell Res 2023; 431:113766. [PMID: 37678504 DOI: 10.1016/j.yexcr.2023.113766] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Roham Deyhimfar
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Sadat Abtahi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrnaz Izady
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Navid Kazemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Atefeh Noori
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
12
|
Tabury K, Rehnberg E, Baselet B, Baatout S, Moroni L. Bioprinting of Cardiac Tissue in Space: Where Are We? Adv Healthc Mater 2023; 12:e2203338. [PMID: 37312654 PMCID: PMC11469151 DOI: 10.1002/adhm.202203338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/18/2023] [Indexed: 06/15/2023]
Abstract
Bioprinting in space is the next frontier in tissue engineering. In the absence of gravity, novel opportunities arise, as well as new challenges. The cardiovascular system needs particular attention in tissue engineering, not only to develop safe countermeasures for astronauts in future deep and long-term space missions, but also to bring solutions to organ transplantation shortage. In this perspective, the challenges encountered when using bioprinting techniques in space and current gaps that need to be overcome are discussed. The recent developments that have been made in the bioprinting of heart tissues in space and an outlook on potential future bioprinting opportunities in space are described.
Collapse
Affiliation(s)
- Kevin Tabury
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
- Department of Biomedical EngineeringCollege of Engineering and ComputingUniversity of South CarolinaColumbiaSC29208USA
| | - Emil Rehnberg
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
- Department of Molecular BiotechnologyGhent UniversityGhent9000Belgium
| | - Bjorn Baselet
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
| | - Sarah Baatout
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
- Department of Molecular BiotechnologyGhent UniversityGhent9000Belgium
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
13
|
Li W, Liu Z, Tang F, Jiang H, Zhou Z, Hao X, Zhang JM. Application of 3D Bioprinting in Liver Diseases. MICROMACHINES 2023; 14:1648. [PMID: 37630184 PMCID: PMC10457767 DOI: 10.3390/mi14081648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
Liver diseases are the primary reason for morbidity and mortality in the world. Owing to a shortage of organ donors and postoperative immune rejection, patients routinely suffer from liver failure. Unlike 2D cell models, animal models, and organoids, 3D bioprinting can be successfully employed to print living tissues and organs that contain blood vessels, bone, and kidney, heart, and liver tissues and so on. 3D bioprinting is mainly classified into four types: inkjet 3D bioprinting, extrusion-based 3D bioprinting, laser-assisted bioprinting (LAB), and vat photopolymerization. Bioinks for 3D bioprinting are composed of hydrogels and cells. For liver 3D bioprinting, hepatic parenchymal cells (hepatocytes) and liver nonparenchymal cells (hepatic stellate cells, hepatic sinusoidal endothelial cells, and Kupffer cells) are commonly used. Compared to conventional scaffold-based approaches, marked by limited functionality and complexity, 3D bioprinting can achieve accurate cell settlement, a high resolution, and more efficient usage of biomaterials, better mimicking the complex microstructures of native tissues. This method will make contributions to disease modeling, drug discovery, and even regenerative medicine. However, the limitations and challenges of this method cannot be ignored. Limitation include the requirement of diverse fabrication technologies, observation of drug dynamic response under perfusion culture, the resolution to reproduce complex hepatic microenvironment, and so on. Despite this, 3D bioprinting is still a promising and innovative biofabrication strategy for the creation of artificial multi-cellular tissues/organs.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Radiology, Yancheng Third People’s Hospital, Affiliated Hospital 6 of Nantong University, Yancheng 224000, China
| | - Zhaoyue Liu
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Fengwei Tang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Hao Jiang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Zhengyuan Zhou
- Nanjing Hangdian Intelligent Manufacturing Technology Co., Ltd., Nanjing 210014, China
| | - Xiuqing Hao
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
| | - Jia Ming Zhang
- College of Mechanical and Electrical Engineering, Nanjing University of Aeronautics and Astronautics; Nanjing 210016, China
- Nanjing Hangdian Intelligent Manufacturing Technology Co., Ltd., Nanjing 210014, China
- Yangtze River Delta Intelligent Manufacturing Innovation Center, Nanjing 210014, China
| |
Collapse
|
14
|
Kutluk H, Bastounis EE, Constantinou I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv Healthc Mater 2023; 12:e2203256. [PMID: 37018430 PMCID: PMC11468608 DOI: 10.1002/adhm.202203256] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/20/2023] [Indexed: 04/07/2023]
Abstract
The extracellular matrix (ECM) is a complex, dynamic network present within all tissues and organs that not only acts as a mechanical support and anchorage point but can also direct fundamental cell behavior, function, and characteristics. Although the importance of the ECM is well established, the integration of well-controlled ECMs into Organ-on-Chip (OoC) platforms remains challenging and the methods to modulate and assess ECM properties on OoCs remain underdeveloped. In this review, current state-of-the-art design and assessment of in vitro ECM environments is discussed with a focus on their integration into OoCs. Among other things, synthetic and natural hydrogels, as well as polydimethylsiloxane (PDMS) used as substrates, coatings, or cell culture membranes are reviewed in terms of their ability to mimic the native ECM and their accessibility for characterization. The intricate interplay among materials, OoC architecture, and ECM characterization is critically discussed as it significantly complicates the design of ECM-related studies, comparability between works, and reproducibility that can be achieved across research laboratories. Improving the biomimetic nature of OoCs by integrating properly considered ECMs would contribute to their further adoption as replacements for animal models, and precisely tailored ECM properties would promote the use of OoCs in mechanobiology.
Collapse
Affiliation(s)
- Hazal Kutluk
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Effie E. Bastounis
- Institute of Microbiology and Infection Medicine (IMIT)Eberhard Karls University of TübingenAuf der Morgenstelle 28, E872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” EXC 2124Eberhard Karls University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
15
|
Stavrou M, Phung N, Grimm J, Andreou C. Organ-on-chip systems as a model for nanomedicine. NANOSCALE 2023; 15:9927-9940. [PMID: 37254663 PMCID: PMC10619891 DOI: 10.1039/d3nr01661g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Nanomedicine is giving rise to increasing numbers of successful drugs, including cancer treatments, molecular imaging agents, and novel vaccine formulations. However, traditionally available model systems offer limited clinical translation and, compared to the number of preclinical studies, the approval rate of nanoparticles (NPs) for clinical use remains disappointingly low. A new paradigm of modeling biological systems on microfluidic chips has emerged in the last decade and is being gradually adopted by the nanomedicine community. These systems mimic tissues, organs, and diseases like cancer, on devices with small physical footprints and complex geometries. In this review, we report studies that used organ-on-chip approaches to study the interactions of NPs with biological systems. We present examples of NP toxicity studies, studies using biological NPs such as viruses, as well as modeling biological barriers and cancer on chip. Organ-on-chip systems present an exciting opportunity and can provide a renewed direction for the nanomedicine community.
Collapse
Affiliation(s)
- Marios Stavrou
- University of Cyprus, Department of Electrical and Computer Engineering, Nicosia, Cyprus.
| | - Ngan Phung
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, New York, NY, USA
- Weill Cornell Medical College, Department of Pharmacology, New York, NY, USA
| | - Jan Grimm
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program, New York, NY, USA
- Weill Cornell Medical College, Department of Pharmacology, New York, NY, USA
| | - Chrysafis Andreou
- University of Cyprus, Department of Electrical and Computer Engineering, Nicosia, Cyprus.
| |
Collapse
|
16
|
Congress Z, Brovold M, Soker S. Cell Viability Assays for 3D Cellular Constructs. Methods Mol Biol 2023; 2644:387-402. [PMID: 37142936 DOI: 10.1007/978-1-0716-3052-5_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
In vitro models fall short of replicating the complex in vivo processes including cell growth and differentiation. For many years, molecular biology research and drug development have relied on the use of cells grown within tissue culture dishes. These traditional in vitro two-dimensional (2D) cultures fail to recapitulate the 3D microenvironment of in vivo tissues. Due to inadequate surface topography, surface stiffness, cell-to-cell, and cell-to-ECM matrices, 2D cell culture systems are incapable of mimicking cell physiology seen in living healthy tissues. These factors can also place selective pressure on cells that substantially alter their molecular and phenotypic properties. With these disadvantages in mind, new and adaptive cell culture systems are necessary to recapitulate the cellular microenvironment in a more accurate manner for drug development, toxicity studies, drug delivery, and much more. Newly developed biofabrication technologies capable of creating 3D tissue constructs can open new opportunities for cell growth and developmental modeling. These constructs show great promise in representing an environment that allows cells to interact with other cells and their microenvironment in a much more physiologically accurate manner. When transitioning from 2D to 3D systems, there is the need to translate common cell viability analysis techniques from that of 2D cell culture to these 3D tissue constructs. Cell viability assays are critical in evaluating the health of cells in response to drug treatment or other stimuli to better understand how these factors effect the tissue constructs. As 3D cellular systems become the new standard in biomedical engineering, this chapter provides different assays used to assess cell viability qualitatively and quantitatively in 3D environments.
Collapse
Affiliation(s)
- Zachary Congress
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
17
|
Chliara MA, Elezoglou S, Zergioti I. Bioprinting on Organ-on-Chip: Development and Applications. BIOSENSORS 2022; 12:1135. [PMID: 36551101 PMCID: PMC9775862 DOI: 10.3390/bios12121135] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Organs-on-chips (OoCs) are microfluidic devices that contain bioengineered tissues or parts of natural tissues or organs and can mimic the crucial structures and functions of living organisms. They are designed to control and maintain the cell- and tissue-specific microenvironment while also providing detailed feedback about the activities that are taking place. Bioprinting is an emerging technology for constructing artificial tissues or organ constructs by combining state-of-the-art 3D printing methods with biomaterials. The utilization of 3D bioprinting and cells patterning in OoC technologies reinforces the creation of more complex structures that can imitate the functions of a living organism in a more precise way. Here, we summarize the current 3D bioprinting techniques and we focus on the advantages of 3D bioprinting compared to traditional cell seeding in addition to the methods, materials, and applications of 3D bioprinting in the development of OoC microsystems.
Collapse
Affiliation(s)
- Maria Anna Chliara
- School of Applied Mathematics and Physical Sciences, National Technical University of Athens, 15780 Zografou, Greece
- Institute of Communication and Computer Systems, 15780 Zografou, Greece
| | - Stavroula Elezoglou
- School of Applied Mathematics and Physical Sciences, National Technical University of Athens, 15780 Zografou, Greece
- PhosPrint P.C., Lefkippos Technology Park, NCSR Demokritos Patriarchou Grigoriou 5’ & Neapoleos 27, 15341 Athens, Greece
| | - Ioanna Zergioti
- School of Applied Mathematics and Physical Sciences, National Technical University of Athens, 15780 Zografou, Greece
- Institute of Communication and Computer Systems, 15780 Zografou, Greece
- PhosPrint P.C., Lefkippos Technology Park, NCSR Demokritos Patriarchou Grigoriou 5’ & Neapoleos 27, 15341 Athens, Greece
| |
Collapse
|
18
|
Unagolla JM, Jayasuriya AC. Recent advances in organoid engineering: A comprehensive review. APPLIED MATERIALS TODAY 2022; 29:101582. [PMID: 38264423 PMCID: PMC10804911 DOI: 10.1016/j.apmt.2022.101582] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Organoid, a 3D structure derived from various cell sources including progenitor and differentiated cells that self-organize through cell-cell and cell-matrix interactions to recapitulate the tissue/organ-specific architecture and function in vitro. The advancement of stem cell culture and the development of hydrogel-based extracellular matrices (ECM) have made it possible to derive self-assembled 3D tissue constructs like organoids. The ability to mimic the actual physiological conditions is the main advantage of organoids, reducing the excessive use of animal models and variability between animal models and humans. However, the complex microenvironment and complex cellular structure of organoids cannot be easily developed only using traditional cell biology. Therefore, several bioengineering approaches, including microfluidics, bioreactors, 3D bioprinting, and organoids-on-a-chip techniques, are extensively used to generate more physiologically relevant organoids. In this review, apart from organoid formation and self-assembly basics, the available bioengineering technologies are extensively discussed as solutions for traditional cell biology-oriented problems in organoid cultures. Also, the natural and synthetic hydrogel systems used in organoid cultures are discussed when necessary to highlight the significance of the stem cell microenvironment. The selected organoid models and their therapeutic applications in drug discovery and disease modeling are also presented.
Collapse
Affiliation(s)
- Janitha M. Unagolla
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, The University of Toledo, Toledo OH, United States
| | - Ambalangodage C. Jayasuriya
- Biomedical Engineering Program, Department of Bioengineering, College of Engineering, The University of Toledo, Toledo OH, United States
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, The University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, United States
| |
Collapse
|
19
|
John P, Antony IR, Whenish R, Jinoop AN. A review on fabrication of 3D printed biomaterials using optical methodologies for tissue engineering applications. Proc Inst Mech Eng H 2022; 236:1583-1594. [DOI: 10.1177/09544119221122856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human body comprises of different internal and external biological components. Human organs tend to fail due to continuous or sudden stress which leads to deterioration, failure, and dislocation. The choice of selection and fabrication of materials for tissue engineering play a key role in terms of suitability, sensitivity, and functioning with other organs as a replacement for failed organs. The progressive improvement of the additive manufacturing (AM) approach in healthcare made it possible to print multi-material and customized complex/intricate geometries in a layer-by-layer fashion. The customized or patient-specific implant fabrication can be easily produced with a high success rate due to the development of AM technologies with tailorable properties. The structural behavior of 3D printed biomaterials is a crucial factor in tissue engineering as they affect the functionality of the implants. Various techniques have been developed in appraising the important features and the effects of the subsequent design of the biomaterial implants. The behavior of the AM built biomaterial implants can be understood visually by an imaging system with a high spatial and spectral resolution. This review intends to present an overview of various biomaterials used in implants, followed by a detailed description of optical 3D printing procedures and evaluation of the performance of 3D printed biomaterials using optical characterization.
Collapse
Affiliation(s)
- Pauline John
- Department of Biomedical Engineering, Sri Sivasubramaniya Nadar College of Engineering, Chennai, TN, India
| | - Irene Rose Antony
- School of Bio-sciences and Technology, Vellore Institute of Technology, Vellore, TN, India
| | - Ruban Whenish
- Center for Biomaterials, Cellular and molecular Theranostics, Vellore Institute of Technology, Vellore, TN, India
| | - Arackal Narayanan Jinoop
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
20
|
Petta D, D'Amora U, D'Arrigo D, Tomasini M, Candrian C, Ambrosio L, Moretti M. Musculoskeletal tissues-on-a-chip: role of natural polymers in reproducing tissue-specific microenvironments. Biofabrication 2022; 14. [PMID: 35931043 DOI: 10.1088/1758-5090/ac8767] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/05/2022] [Indexed: 11/12/2022]
Abstract
Over the past years, 3D in vitro models have been widely employed in the regenerative medicine field. Among them, organ-on-a-chip technology has the potential to elucidate cellular mechanism exploiting multichannel microfluidic devices to establish 3D co-culture systems that offer control over the cellular, physico-chemical and biochemical microenvironments. To deliver the most relevant cues to cells, it is of paramount importance to select the most appropriate matrix for mimicking the extracellular matrix of the native tissue. Natural polymers-based hydrogels are the elected candidates for reproducing tissue-specific microenvironments in musculoskeletal tissue-on-a-chip models owning to their interesting and peculiar physico-chemical, mechanical and biological properties. Despite these advantages, there is still a gap between the biomaterials complexity in conventional tissue engineering and the application of these biomaterials in 3D in vitro microfluidic models. In this review, the aim is to suggest the adoption of more suitable biomaterials, alternative crosslinking strategies and tissue engineered-inspired approaches in organ-on-a-chip to better mimic the complexity of physiological musculoskeletal tissues. Accordingly, after giving an overview of the musculoskeletal tissue compositions, the properties of the main natural polymers employed in microfluidic systems are investigated, together with the main musculoskeletal tissues-on-a-chip devices.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologis Lab, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Ugo D'Amora
- Institute of Polymers, Composites and Biomaterials, National Research Council, V.le J.F. Kennedy 54 Mostra d'Oltremare Pad 20, Naples, 80125, ITALY
| | - Daniele D'Arrigo
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Marta Tomasini
- Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco chies 5, Bellinzona, Ticino, 6500, SWITZERLAND
| | - Christian Candrian
- Unità di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale, via Tesserete 46, Lugano, 6900, SWITZERLAND
| | - Luigi Ambrosio
- Institute of Polymers Composites and Biomaterials National Research Council, Viale Kennedy, Pozzuoli, Campania, 80078, ITALY
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Repubblica e Cantone Ticino Ente Ospedaliero Cantonale, Via Francesco Chiesa 5, Bellinzona, Ticino, 6500, SWITZERLAND
| |
Collapse
|
21
|
Abstract
Drug testing, either on animals or on 2D cell cultures, has its limitations due to inaccurate mimicking of human pathophysiology. The liver, as one of the key organs that filters and detoxifies the blood, is susceptible to drug-induced injuries. Integrating 3D bioprinting with microfluidic chips to fabricate organ-on-chip platforms for 3D liver cell cultures with continuous perfusion can offer a more physiologically relevant liver-mimetic platform for screening drugs and studying liver function. The development of organ-on-chip platforms may ultimately contribute to personalized medicine as well as body-on-chip technology that can test drug responses and organ–organ interactions on a single or linked chip model.
Collapse
|
22
|
Kavand H, Nasiri R, Herland A. Advanced Materials and Sensors for Microphysiological Systems: Focus on Electronic and Electrooptical Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107876. [PMID: 34913206 DOI: 10.1002/adma.202107876] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Advanced in vitro cell culture systems or microphysiological systems (MPSs), including microfluidic organ-on-a-chip (OoC), are breakthrough technologies in biomedicine. These systems recapitulate features of human tissues outside of the body. They are increasingly being used to study the functionality of different organs for applications such as drug evolutions, disease modeling, and precision medicine. Currently, developers and endpoint users of these in vitro models promote how they can replace animal models or even be a better ethically neutral and humanized alternative to study pathology, physiology, and pharmacology. Although reported models show a remarkable physiological structure and function compared to the conventional 2D cell culture, they are almost exclusively based on standard passive polymers or glass with none or minimal real-time stimuli and readout capacity. The next technology leap in reproducing in vivo-like functionality and real-time monitoring of tissue function could be realized with advanced functional materials and devices. This review describes the currently reported electronic and optical advanced materials for sensing and stimulation of MPS models. In addition, an overview of multi-sensing for Body-on-Chip platforms is given. Finally, one gives the perspective on how advanced functional materials could be integrated into in vitro systems to precisely mimic human physiology.
Collapse
Affiliation(s)
- Hanie Kavand
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
| | - Rohollah Nasiri
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| | - Anna Herland
- Division of Micro- and Nanosystems, Department of Intelligent Systems, KTH Royal Institute of Technology, Malvinas Väg 10 pl 5, Stockholm, 100 44, Sweden
- AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solnavägen 9/B8, Solna, 171 65, Sweden
- Division of Nanobiotechnology, Department of Protein Science, KTH Royal Institute of Technology, Tomtebodavägen 23a, Solna, 171 65, Sweden
| |
Collapse
|
23
|
Nadine S, Chung A, Diltemiz SE, Yasuda B, Lee C, Hosseini V, Karamikamkar S, de Barros NR, Mandal K, Advani S, Zamanian BB, Mecwan M, Zhu Y, Mofidfar M, Zare MR, Mano J, Dokmeci MR, Alambeigi F, Ahadian S. Advances in microfabrication technologies in tissue engineering and regenerative medicine. Artif Organs 2022; 46:E211-E243. [PMID: 35349178 DOI: 10.1111/aor.14232] [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: 01/14/2022] [Revised: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tissue engineering provides various strategies to fabricate an appropriate microenvironment to support the repair and regeneration of lost or damaged tissues. In this matter, several technologies have been implemented to construct close-to-native three-dimensional structures at numerous physiological scales, which are essential to confer the functional characteristics of living tissues. METHODS In this article, we review a variety of microfabrication technologies that are currently utilized for several tissue engineering applications, such as soft lithography, microneedles, templated and self-assembly of microstructures, microfluidics, fiber spinning, and bioprinting. RESULTS These technologies have considerably helped us to precisely manipulate cells or cellular constructs for the fabrication of biomimetic tissues and organs. Although currently available tissues still lack some crucial functionalities, including vascular networks, innervation, and lymphatic system, microfabrication strategies are being proposed to overcome these issues. Moreover, the microfabrication techniques that have progressed to the preclinical stage are also discussed. CONCLUSIONS This article aims to highlight the advantages and drawbacks of each technique and areas of further research for a more comprehensive and evolving understanding of microfabrication techniques in terms of tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Sara Nadine
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA.,CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Ada Chung
- Department of Psychology, University of California-Los Angeles, Los Angeles, California, USA
| | | | - Brooke Yasuda
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA.,Department of Psychology, University of California-Los Angeles, Los Angeles, California, USA
| | - Charles Lee
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA.,Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, USA.,Station 1, Lawrence, Massachusetts, USA
| | - Vahid Hosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Solmaz Karamikamkar
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | | | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Shailesh Advani
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | | | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Mohammad Mofidfar
- Department of Chemistry, Stanford University, Palo Alto, California, USA
| | | | - João Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Farshid Alambeigi
- Walker Department of Mechanical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| |
Collapse
|
24
|
Shukla AK, Gao G, Kim BS. Applications of 3D Bioprinting Technology in Induced Pluripotent Stem Cells-Based Tissue Engineering. MICROMACHINES 2022; 13:155. [PMID: 35208280 PMCID: PMC8876961 DOI: 10.3390/mi13020155] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are essentially produced by the genetic reprogramming of adult cells. Moreover, iPSC technology prevents the genetic manipulation of embryos. Hence, with the ensured element of safety, they rarely cause ethical concerns when utilized in tissue engineering. Several cumulative outcomes have demonstrated the functional superiority and potency of iPSCs in advanced regenerative medicine. Recently, an emerging trend in 3D bioprinting technology has been a more comprehensive approach to iPSC-based tissue engineering. The principal aim of this review is to provide an understanding of the applications of 3D bioprinting in iPSC-based tissue engineering. This review discusses the generation of iPSCs based on their distinct purpose, divided into two categories: (1) undifferentiated iPSCs applied with 3D bioprinting; (2) differentiated iPSCs applied with 3D bioprinting. Their significant potential is analyzed. Lastly, various applications for engineering tissues and organs have been introduced and discussed in detail.
Collapse
Affiliation(s)
- Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Korea;
| | - Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China
- Department of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Korea;
| |
Collapse
|
25
|
Imparato G, Urciuolo F, Netti PA. Organ on Chip Technology to Model Cancer Growth and Metastasis. Bioengineering (Basel) 2022; 9:28. [PMID: 35049737 PMCID: PMC8772984 DOI: 10.3390/bioengineering9010028] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/18/2022] Open
Abstract
Organ on chip (OOC) has emerged as a major technological breakthrough and distinct model system revolutionizing biomedical research and drug discovery by recapitulating the crucial structural and functional complexity of human organs in vitro. OOC are rapidly emerging as powerful tools for oncology research. Indeed, Cancer on chip (COC) can ideally reproduce certain key aspects of the tumor microenvironment (TME), such as biochemical gradients and niche factors, dynamic cell-cell and cell-matrix interactions, and complex tissue structures composed of tumor and stromal cells. Here, we review the state of the art in COC models with a focus on the microphysiological systems that host multicellular 3D tissue engineering models and can help elucidate the complex biology of TME and cancer growth and progression. Finally, some examples of microengineered tumor models integrated with multi-organ microdevices to study disease progression in different tissues will be presented.
Collapse
Affiliation(s)
- Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; (F.U.); (P.A.N.)
| | - Francesco Urciuolo
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Department of Chemical, Materials and Industrial Production (DICMAPI), Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.leTecchio 80, 80125 Naples, Italy
| | - Paolo Antonio Netti
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Department of Chemical, Materials and Industrial Production (DICMAPI), Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.leTecchio 80, 80125 Naples, Italy
| |
Collapse
|
26
|
Samanipour R, Tahmooressi H, Rezaei Nejad H, Hirano M, Shin SR, Hoorfar M. A review on 3D printing functional brain model. BIOMICROFLUIDICS 2022; 16:011501. [PMID: 35145569 PMCID: PMC8816519 DOI: 10.1063/5.0074631] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/31/2021] [Indexed: 05/08/2023]
Abstract
Modern neuroscience increasingly relies on 3D models to study neural circuitry, nerve regeneration, and neural disease. Several different biofabrication approaches have been explored to create 3D neural tissue model structures. Among them, 3D bioprinting has shown to have great potential to emerge as a high-throughput/high precision biofabrication strategy that can address the growing need for 3D neural models. Here, we have reviewed the design principles for neural tissue engineering. The main challenge to adapt printing technologies for biofabrication of neural tissue models is the development of neural bioink, i.e., a biomaterial with printability and gelation properties and also suitable for neural tissue culture. This review shines light on a vast range of biomaterials as well as the fundamentals of 3D neural tissue printing. Also, advances in 3D bioprinting technologies are reviewed especially for bioprinted neural models. Finally, the techniques used to evaluate the fabricated 2D and 3D neural models are discussed and compared in terms of feasibility and functionality.
Collapse
Affiliation(s)
| | - Hamed Tahmooressi
- Department of Mechanical Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Hojatollah Rezaei Nejad
- Department of Electrical and Computer Engineering, Tufts University, 161 College Avenue, Medford, Massachusetts 02155, USA
| | | | - Su-Royn Shin
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02139, USA
- Authors to whom correspondence should be addressed: and
| | - Mina Hoorfar
- Faculty of Engineering, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
27
|
Taghizadeh M, Taghizadeh A, Yazdi MK, Zarrintaj P, Stadler FJ, Ramsey JD, Habibzadeh S, Hosseini Rad S, Naderi G, Saeb MR, Mozafari M, Schubert US. Chitosan-based inks for 3D printing and bioprinting. GREEN CHEMISTRY 2022; 24:62-101. [DOI: 10.1039/d1gc01799c] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
3D printing gave biomedical engineering great potential to mimic native tissues, accelerated regenerative medicine, and enlarged capacity of drug delivery systems; thus, advanced biomimetic functional biomaterial developed by 3D-printing for tissue engineering demands.
Collapse
Affiliation(s)
- Mohsen Taghizadeh
- College of Materials Science and Engineering, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, Shenzhen University, Shenzhen 518060, PR China
| | - Ali Taghizadeh
- College of Materials Science and Engineering, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, Shenzhen University, Shenzhen 518060, PR China
| | - Mohsen Khodadadi Yazdi
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, USA
| | - Florian J. Stadler
- College of Materials Science and Engineering, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, Shenzhen University, Shenzhen 518060, PR China
| | - Joshua D. Ramsey
- School of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK 74078, USA
| | - Sajjad Habibzadeh
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 15916-39675, Iran
| | - Somayeh Hosseini Rad
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC, H3C 3A7, Canada
| | - Ghasem Naderi
- Iran Polymer and Petrochemical Institute (IPPI), Tehran, Iran
| | - Mohammad Reza Saeb
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11, /12 80-233, Gdańsk, Poland
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Center for Energy and Environmental Chemistry Jena (CEEC Jena), Friedrich Schiller University Jena, Philosophenweg 7a, 07743, Jena, Germany
| |
Collapse
|
28
|
Jamee R, Araf Y, Naser IB, Promon SK. The promising rise of bioprinting in revolutionalizing medical science: Advances and possibilities. Regen Ther 2021; 18:133-145. [PMID: 34189195 PMCID: PMC8213915 DOI: 10.1016/j.reth.2021.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 12/24/2022] Open
Abstract
Bioprinting is a relatively new yet evolving technique predominantly used in regenerative medicine and tissue engineering. 3D bioprinting techniques combine the advantages of creating Extracellular Matrix (ECM)like environments for cells and computer-aided tailoring of predetermined tissue shapes and structures. The essential application of bioprinting is for the regeneration or restoration of damaged and injured tissues by producing implantable tissues and organs. The capability of bioprinting is yet to be fully scrutinized in sectors like the patient-specific spatial distribution of cells, bio-robotics, etc. In this review, currently developed experimental systems and strategies for the bioprinting of different types of tissues as well as for drug delivery and cancer research are explored for potential applications. This review also digs into the most recent opportunities and future possibilities for the efficient implementation of bioprinting to restructure medical and technological practices.
Collapse
Affiliation(s)
- Radia Jamee
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
- Mechamind, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Iftekhar Bin Naser
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
| | - Salman Khan Promon
- Department of Mathematics and Natural Sciences, School of Data and Sciences, Brac University, Dhaka, Bangladesh
- Mechamind, Dhaka, Bangladesh
| |
Collapse
|
29
|
Dellaquila A, Le Bao C, Letourneur D, Simon‐Yarza T. In Vitro Strategies to Vascularize 3D Physiologically Relevant Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100798. [PMID: 34351702 PMCID: PMC8498873 DOI: 10.1002/advs.202100798] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/23/2021] [Indexed: 05/04/2023]
Abstract
Vascularization of 3D models represents a major challenge of tissue engineering and a key prerequisite for their clinical and industrial application. The use of prevascularized models built from dedicated materials could solve some of the actual limitations, such as suboptimal integration of the bioconstructs within the host tissue, and would provide more in vivo-like perfusable tissue and organ-specific platforms. In the last decade, the fabrication of vascularized physiologically relevant 3D constructs has been attempted by numerous tissue engineering strategies, which are classified here in microfluidic technology, 3D coculture models, namely, spheroids and organoids, and biofabrication. In this review, the recent advancements in prevascularization techniques and the increasing use of natural and synthetic materials to build physiological organ-specific models are discussed. Current drawbacks of each technology, future perspectives, and translation of vascularized tissue constructs toward clinics, pharmaceutical field, and industry are also presented. By combining complementary strategies, these models are envisioned to be successfully used for regenerative medicine and drug development in a near future.
Collapse
Affiliation(s)
- Alessandra Dellaquila
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Elvesys Microfluidics Innovation CenterParis75011France
- Biomolecular PhotonicsDepartment of PhysicsUniversity of BielefeldBielefeld33615Germany
| | - Chau Le Bao
- Université de ParisINSERM U1148X Bichat HospitalParisF‐75018France
- Université Sorbonne Paris NordGalilée InstituteVilletaneuseF‐93430France
| | | | | |
Collapse
|
30
|
Malik M, Yang Y, Fathi P, Mahler GJ, Esch MB. Critical Considerations for the Design of Multi-Organ Microphysiological Systems (MPS). Front Cell Dev Biol 2021; 9:721338. [PMID: 34568333 PMCID: PMC8459628 DOI: 10.3389/fcell.2021.721338] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Identification and approval of new drugs for use in patients requires extensive preclinical studies and clinical trials. Preclinical studies rely on in vitro experiments and animal models of human diseases. The transferability of drug toxicity and efficacy estimates to humans from animal models is being called into question. Subsequent clinical studies often reveal lower than expected efficacy and higher drug toxicity in humans than that seen in animal models. Microphysiological systems (MPS), sometimes called organ or human-on-chip models, present a potential alternative to animal-based models used for drug toxicity screening. This review discusses multi-organ MPS that can be used to model diseases and test the efficacy and safety of drug candidates. The translation of an in vivo environment to an in vitro system requires physiologically relevant organ scaling, vascular dimensions, and appropriate flow rates. Even small changes in those parameters can alter the outcome of experiments conducted with MPS. With many MPS devices being developed, we have outlined some established standards for designing MPS devices and described techniques to validate the devices. A physiologically realistic mimic of the human body can help determine the dose response and toxicity effects of a new drug candidate with higher predictive power.
Collapse
Affiliation(s)
- Mridu Malik
- Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| | - Yang Yang
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
- Department of Chemical Engineering, University of Maryland, College Park, College Park, MD, United States
| | - Parinaz Fathi
- Department of Bioengineering, Materials Science and Engineering, and Beckman Institute, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Gretchen J. Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
| | - Mandy B. Esch
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| |
Collapse
|
31
|
Bioprinting of Organ-on-Chip Systems: A Literature Review from a Manufacturing Perspective. JOURNAL OF MANUFACTURING AND MATERIALS PROCESSING 2021. [DOI: 10.3390/jmmp5030091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review discusses the reported studies investigating the use of bioprinting to develop functional organ-on-chip systems from a manufacturing perspective. These organ-on-chip systems model the liver, kidney, heart, lung, gut, bone, vessel, and tumors to demonstrate the viability of bioprinted organ-on-chip systems for disease modeling and drug screening. In addition, the paper highlights the challenges involved in using bioprinting techniques for organ-on-chip system fabrications and suggests future research directions. Based on the reviewed studies, it is concluded that bioprinting can be applied for the automated and assembly-free fabrication of organ-on chip systems. These bioprinted organ-on-chip systems can help in the modeling of several different diseases and can thereby expedite drug discovery by providing an efficient platform for drug screening in the preclinical phase of drug development processes.
Collapse
|
32
|
Elalouf A. Immune response against the biomaterials used in 3D bioprinting of organs. Transpl Immunol 2021; 69:101446. [PMID: 34389430 DOI: 10.1016/j.trim.2021.101446] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 12/26/2022]
Abstract
Regenerative medicine has developed promising approaches for healing and replacing defective and damaged organs or tissues with functional ones. Three-dimensional (3D) bioprinting innovation has integrated a potential to design organs or tissues specific to the patient with the capability of rapid construction to fulfill the storage of organs and the need for transplantation. 3D bioprinting of organs has the main goal to develop a structural and functional organ or tissue mimic to the original one. The highly complex fabrication of tissue engineering scaffolds containing biomaterials, tissue models, and biomedical devices has made it possible to print small blood vessels to mimic organs to reduce organ or tissue rejection. 3D bioprinting has the concept of bioinks containing biomaterials that may trigger the immune responses in the body. Nevertheless, foreign body response (FBR) is mediated by various cell types such as B-cells, dendritic cells, macrophages, natural killer cells, neutrophils, and T-cells, and molecular signals such as antibodies (Abs), cytokines, and reactive radical species. Typically, the biomaterial is shielded by the fibrous encapsulation that is regulated by molecular signals. This review explored the progress in 3D bioprinting of vital organs and basic immune response against the biomaterials used in this approach. Thus, evaluating immune response against biomaterials used in 3D printed organs is necessary to mitigate tissue rejection after the transplantation.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| |
Collapse
|
33
|
Use of electroconductive biomaterials for engineering tissues by 3D printing and 3D bioprinting. Essays Biochem 2021; 65:441-466. [PMID: 34296738 DOI: 10.1042/ebc20210003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Existing methods of engineering alternatives to restore or replace damaged or lost tissues are not satisfactory due to the lack of suitable constructs that can fit precisely, function properly and integrate into host tissues. Recently, three-dimensional (3D) bioprinting approaches have been developed to enable the fabrication of pre-programmed synthetic tissue constructs that have precise geometries and controlled cellular composition and spatial distribution. New bioinks with electroconductive properties have the potential to influence cellular fates and function for directed healing of different tissue types including bone, heart and nervous tissue with the possibility of improved outcomes. In the present paper, we review the use of electroconductive biomaterials for the engineering of tissues via 3D printing and 3D bioprinting. Despite significant advances, there remain challenges to effective tissue replacement and we address these challenges and describe new approaches to advanced tissue engineering.
Collapse
|
34
|
Tavafoghi M, Darabi MA, Mahmoodi M, Tutar R, Xu C, Mirjafari A, Billi F, Swieszkowski W, Nasrollahi F, Ahadian S, Hosseini V, Khademhosseini A, Ashammakhi N. Multimaterial bioprinting and combination of processing techniques towards the fabrication of biomimetic tissues and organs. Biofabrication 2021; 13. [PMID: 34130266 DOI: 10.1088/1758-5090/ac0b9a] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022]
Abstract
Tissue reconstruction requires the utilization of multiple biomaterials and cell types to replicate the delicate and complex structure of native tissues. Various three-dimensional (3D) bioprinting techniques have been developed to fabricate customized tissue structures; however, there are still significant challenges, such as vascularization, mechanical stability of printed constructs, and fabrication of gradient structures to be addressed for the creation of biomimetic and complex tissue constructs. One approach to address these challenges is to develop multimaterial 3D bioprinting techniques that can integrate various types of biomaterials and bioprinting capabilities towards the fabrication of more complex structures. Notable examples include multi-nozzle, coaxial, and microfluidics-assisted multimaterial 3D bioprinting techniques. More advanced multimaterial 3D printing techniques are emerging, and new areas in this niche technology are rapidly evolving. In this review, we briefly introduce the basics of individual 3D bioprinting techniques and then discuss the multimaterial 3D printing techniques that can be developed based on combination of these techniques for the engineering of complex and biomimetic tissue constructs. We also discuss the perspectives and future directions to develop state-of-the-art multimaterial 3D bioprinting techniques for engineering tissues and organs.
Collapse
Affiliation(s)
- Maryam Tavafoghi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Mahboobeh Mahmoodi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Rumeysa Tutar
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa Avcılar, Istanbul 34320, Turkey
| | - Chun Xu
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,School of Dentistry, The University of Queensland, Brisbane, Australia
| | - Arshia Mirjafari
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America
| | - Fabrizio Billi
- UCLA/OIC Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States of America
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Fatemeh Nasrollahi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Vahid Hosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Bioengineering, University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Terasaki Institute for Biomedical Innovation, Los Angeles, CA, United States of America.,Department of Chemical Engineering, University of California, Los Angeles, CA, United States of America
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, United States of America.,Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America.,Department of Biomedical Engineering, College of Engineering, Michigan State University, MI, United States of America
| |
Collapse
|
35
|
Moghaddam AS, Khonakdar HA, Arjmand M, Jafari SH, Bagher Z, Moghaddam ZS, Chimerad M, Sisakht MM, Shojaei S. Review of Bioprinting in Regenerative Medicine: Naturally Derived Bioinks and Stem Cells. ACS APPLIED BIO MATERIALS 2021; 4:4049-4070. [PMID: 35006822 DOI: 10.1021/acsabm.1c00219] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative medicine offers the potential to repair or substitute defective tissues by constructing active tissues to address the scarcity and demands for transplantation. The method of forming 3D constructs made up of biomaterials, cells, and biomolecules is called bioprinting. Bioprinting of stem cells provides the ability to reliably recreate tissues, organs, and microenvironments to be used in regenerative medicine. 3D bioprinting is a technique that uses several biomaterials and cells to tailor a structure with clinically relevant geometries and sizes. This technique's promise is demonstrated by 3D bioprinted tissues, including skin, bone, cartilage, and cardiovascular, corneal, hepatic, and adipose tissues. Several bioprinting methods have been combined with stem cells to effectively produce tissue models, including adult stem cells, embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and differentiation techniques. In this review, technological challenges of printed stem cells using prevalent naturally derived bioinks (e.g., carbohydrate polymers and protein-based polymers, peptides, and decellularized extracellular matrix), recent advancements, leading companies, and clinical trials in the field of 3D bioprinting are delineated.
Collapse
Affiliation(s)
- Abolfazl Salehi Moghaddam
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4593, Iran
| | - Hossein Ali Khonakdar
- Leibniz Institute of Polymer Research Dresden, Hohe Straße 6, Dresden D-01069, Germany.,Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
| | - Mohammad Arjmand
- Nanomaterials and Polymer Nanocomposites Laboratory, School of Engineering, University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Seyed Hassan Jafari
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4593, Iran
| | - Zohreh Bagher
- ENT and Head & Neck Research Centre and Department, The Five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Zahra Salehi Moghaddam
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Mohammadreza Chimerad
- School of Mechanical Engineering, Iran University of Science and Technology, Tehran 16844, Iran
| | - Mahsa Mollapour Sisakht
- Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran 19379-57511, Iran.,Department of Biochemistry, Erasmus University Medical Center, Rotterdam 3000 DR, The Netherlands
| | - Shahrokh Shojaei
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, PO Box 13185/768, Tehran 15689-37813, Iran.,Stem Cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Tehran Branch, PO Box 13185-768, Tehran 15689-37813, Iran
| |
Collapse
|
36
|
Ghazimirsaeed E, Madadelahi M, Dizani M, Shamloo A. Secondary Flows, Mixing, and Chemical Reaction Analysis of Droplet-Based Flow inside Serpentine Microchannels with Different Cross Sections. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:5118-5130. [PMID: 33877832 DOI: 10.1021/acs.langmuir.0c03662] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Chemical bioreactions are an important aspect of many recent microfluidic devices, and their applications in biomedical science have been growing worldwide. Droplet-based microreactors are among the attractive types of unit operations, which utilize droplets for enhancement in both mixing and chemical reactions. In the present study, a finite-volume-method (FVM) numerical investigation is conducted based on the volume-of-fluid (VOF) applying for the droplet-based flows. This multiphase computational modeling is used for the study of the chemical reaction and mixing phenomenon inside a serpentine microchannel and explores the effects of the aspect ratio (i.e., AR = height/width) of rectangular cross-sectional geometries as well as three other cross-sectional geometries including trapezoidal, triangular, and circular, on consumption and production rates of chemical species. It is found that in these droplet bioreactors, the reaction begins from the forward section of the droplet. We investigate the secondary flows and chemical reactions inside the droplets in a serpentine microchannel with different cross-sectional geometries. Different transient Dean vortices and secondary flows in the presence and absence of the droplets are studied and explained based on the position of the droplets. It is found that as the droplets pass through the microchannel turns, the patterns and magnitude of the secondary flows change, depending on the cross-sectional geometry. Eventually, the results demonstrate that the AR = 2 rectangular cross-section is the most helpful geometry, whereas the trapezoidal cross-section takes into account the least efficient one between all geometries.
Collapse
Affiliation(s)
- Erfan Ghazimirsaeed
- Department of Mechanical Engineering, Sharif University of Technology, >Tehran 165165161, Iran
| | - Masoud Madadelahi
- Department of Mechanical Engineering, Sharif University of Technology, >Tehran 165165161, Iran
| | - Mahdi Dizani
- Department of Mechanical Engineering, Sharif University of Technology, >Tehran 165165161, Iran
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, >Tehran 165165161, Iran
| |
Collapse
|
37
|
Jacob S, Nair AB, Shah J, Sreeharsha N, Gupta S, Shinu P. Emerging Role of Hydrogels in Drug Delivery Systems, Tissue Engineering and Wound Management. Pharmaceutics 2021; 13:357. [PMID: 33800402 PMCID: PMC7999964 DOI: 10.3390/pharmaceutics13030357] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/27/2021] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
The popularity of hydrogels as biomaterials lies in their tunable physical properties, ability to encapsulate small molecules and macromolecular drugs, water holding capacity, flexibility, and controllable degradability. Functionalization strategies to overcome the deficiencies of conventional hydrogels and expand the role of advanced hydrogels such as DNA hydrogels are extensively discussed in this review. Different types of cross-linking techniques, materials utilized, procedures, advantages, and disadvantages covering hydrogels are tabulated. The application of hydrogels, particularly in buccal, oral, vaginal, and transdermal drug delivery systems, are described. The review also focuses on composite hydrogels with enhanced properties that are being developed to meet the diverse demand of wound dressing materials. The unique advantages of hydrogel nanoparticles in targeted and intracellular delivery of various therapeutic agents are explained. Furthermore, different types of hydrogel-based materials utilized for tissue engineering applications and fabrication of contact lens are discussed. The article also provides an overview of selected examples of commercial products launched particularly in the area of oral and ocular drug delivery systems and wound dressing materials. Hydrogels can be prepared with a wide variety of properties, achieving biostable, bioresorbable, and biodegradable polymer matrices, whose mechanical properties and degree of swelling are tailored with a specific application. These unique features give them a promising future in the fields of drug delivery systems and applied biomedicine.
Collapse
Affiliation(s)
- Shery Jacob
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Anroop B. Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (N.S.)
| | - Jigar Shah
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India;
| | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (A.B.N.); (N.S.)
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bangalore 560035, India
| | - Sumeet Gupta
- Department of Pharmacology, M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana 133203, India;
| | - Pottathil Shinu
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| |
Collapse
|
38
|
In Vitro Human Joint Models Combining Advanced 3D Cell Culture and Cutting-Edge 3D Bioprinting Technologies. Cells 2021; 10:cells10030596. [PMID: 33800436 PMCID: PMC7999996 DOI: 10.3390/cells10030596] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
Joint-on-a-chip is a new technology able to replicate the joint functions into microscale systems close to pathophysiological conditions. Recent advances in 3D printing techniques allow the precise control of the architecture of the cellular compartments (including chondrocytes, stromal cells, osteocytes and synoviocytes). These tools integrate fluid circulation, the delivery of growth factors, physical stimulation including oxygen level, external pressure, and mobility. All of these structures must be able to mimic the specific functions of the diarthrodial joint: mobility, biomechanical aspects and cellular interactions. All the elements must be grouped together in space and reorganized in a manner close to the joint organ. This will allow the study of rheumatic disease physiopathology, the development of biomarkers and the screening of new drugs.
Collapse
|
39
|
Picollet-D'hahan N, Zuchowska A, Lemeunier I, Le Gac S. Multiorgan-on-a-Chip: A Systemic Approach To Model and Decipher Inter-Organ Communication. Trends Biotechnol 2021; 39:788-810. [PMID: 33541718 DOI: 10.1016/j.tibtech.2020.11.014] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
Multiorgan-on-a-chip (multi-OoC) platforms have great potential to redefine the way in which human health research is conducted. After briefly reviewing the need for comprehensive multiorgan models with a systemic dimension, we highlight scenarios in which multiorgan models are advantageous. We next overview existing multi-OoC platforms, including integrated body-on-a-chip devices and modular approaches involving interconnected organ-specific modules. We highlight how multi-OoC models can provide unique information that is not accessible using single-OoC models. Finally, we discuss remaining challenges for the realization of multi-OoC platforms and their worldwide adoption. We anticipate that multi-OoC technology will metamorphose research in biology and medicine by providing holistic and personalized models for understanding and treating multisystem diseases.
Collapse
Affiliation(s)
- Nathalie Picollet-D'hahan
- Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale (INSERM), Commissariat à l'Energie Atomique (CEA) Interdisciplinary Research Institute of Grenoble (IRIG) Biomicrotechnology and Functional Genomics (BIOMICS), Grenoble, France.
| | - Agnieszka Zuchowska
- Applied Microfluidics for Bioengineering Research (AMBER), MESA+ Institute for Nanotechnology, TechMed Center, University of Twente, 7500AE Enschede, The Netherlands
| | - Iris Lemeunier
- Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale (INSERM), Commissariat à l'Energie Atomique (CEA) Interdisciplinary Research Institute of Grenoble (IRIG) Biomicrotechnology and Functional Genomics (BIOMICS), Grenoble, France
| | - Séverine Le Gac
- Applied Microfluidics for Bioengineering Research (AMBER), MESA+ Institute for Nanotechnology, TechMed Center, University of Twente, 7500AE Enschede, The Netherlands.
| |
Collapse
|
40
|
Kjar A, McFarland B, Mecham K, Harward N, Huang Y. Engineering of tissue constructs using coaxial bioprinting. Bioact Mater 2021; 6:460-471. [PMID: 32995673 PMCID: PMC7490764 DOI: 10.1016/j.bioactmat.2020.08.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/12/2020] [Accepted: 08/23/2020] [Indexed: 12/13/2022] Open
Abstract
Bioprinting is a rapidly developing technology for the precise design and manufacture of tissues in various biological systems or organs. Coaxial extrusion bioprinting, an emergent branch, has demonstrated a strong potential to enhance bioprinting's engineering versatility. Coaxial bioprinting assists in the fabrication of complex tissue constructs, by enabling concentric deposition of biomaterials. The fabricated tissue constructs started with simple, tubular vasculature but have been substantially developed to integrate complex cell composition and self-assembly, ECM patterning, controlled release, and multi-material gradient profiles. This review article begins with a brief overview of coaxial printing history, followed by an introduction of crucial engineering components. Afterward, we review the recent progress and untapped potential in each specific organ or biological system, and demonstrate how coaxial bioprinting facilitates the creation of tissue constructs. Ultimately, we conclude that this growing technology will contribute significantly to capabilities in the fields of in vitro modeling, pharmaceutical development, and clinical regenerative medicine.
Collapse
Affiliation(s)
- Andrew Kjar
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| | - Bailey McFarland
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| | - Keetch Mecham
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| | - Nathan Harward
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| | - Yu Huang
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| |
Collapse
|
41
|
Rasheed A, Azizi L, Turkki P, Janka M, Hytönen VP, Tuukkanen S. Extrusion-Based Bioprinting of Multilayered Nanocellulose Constructs for Cell Cultivation Using In Situ Freezing and Preprint CaCl 2 Cross-Linking. ACS OMEGA 2021; 6:569-578. [PMID: 33458509 PMCID: PMC7807796 DOI: 10.1021/acsomega.0c05036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/11/2020] [Indexed: 05/05/2023]
Abstract
Extrusion-based bioprinting with a preprint cross-linking agent and an in situ cooling stage provides a versatile method for the fabrication of 3D structures for cell culture. We added varying amounts of calcium chloride as a precross-linker into native nanofibrillated cellulose (NFC) hydrogel prior to 3D bioprinting to fabricate structurally stable multilayered constructs without the need for a separate cross-linking bath. To further enhance their stability, we bioprinted the multilayered structures onto an in situ temperature-controlled printing stage at 25, 0, and -10 °C. The extruded and subsequently freeze-dried volumetric constructs maintained their structures after being immersed into a cell culture medium. The ability to maintain the shape after immersion in cell media is an essential feature for the fabrication of stem cell-based artificial organs. We studied the viability and distribution of mouse embryonic fibroblast cells into the hydrogels using luminescence technique and confocal microscopy. Adding CaCl2 increased the stability of the multilayered nanocellulose structures, making them suitable for culturing cells inside the 3D hydrogel environment. Lower stage temperature considerably improved the structural stability of the 3D printed structures, however, had no effect on cell viability.
Collapse
Affiliation(s)
- Anum Rasheed
- Faculty
of Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 7 Kampusareena, 33720 Tampere, Finland
| | - Latifeh Azizi
- Faculty
of Medicine and Health Technology, Tampere
University, Arvo Ylpön
Katu 34, 33520 Tampere, Finland
| | - Paula Turkki
- Faculty
of Medicine and Health Technology, Tampere
University, Arvo Ylpön
Katu 34, 33520 Tampere, Finland
| | - Marika Janka
- Faculty
of Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 7 Kampusareena, 33720 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty
of Medicine and Health Technology, Tampere
University, Arvo Ylpön
Katu 34, 33520 Tampere, Finland
- Fimlab
Laboratories, Biokatu
4, 33520 Tampere, Finland
| | - Sampo Tuukkanen
- Faculty
of Medicine and Health Technology, Tampere
University, Korkeakoulunkatu 7 Kampusareena, 33720 Tampere, Finland
| |
Collapse
|
42
|
Fritschen A, Blaeser A. Biosynthetic, biomimetic, and self-assembled vascularized Organ-on-a-Chip systems. Biomaterials 2020; 268:120556. [PMID: 33310539 DOI: 10.1016/j.biomaterials.2020.120556] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Organ-on-a-Chip (OOC) devices have seen major advances in the last years with respect to biological complexity, physiological composition and biomedical relevance. In this context, integration of vasculature has proven to be a crucial element for long-term culture of thick tissue samples as well as for realistic pharmacokinetic, toxicity and metabolic modelling. With the emergence of digital production technologies and the reinvention of existing tools, a multitude of design approaches for guided angio- and vasculogenesis is available today. The underlying production methods can be categorized into biosynthetic, biomimetic and self-assembled vasculature formation. The diversity and importance of production approaches, vascularization strategies as well as biomaterials and cell sourcing are illustrated in this work. A comprehensive technological review with a strong focus on the challenge of producing physiologically relevant vascular structures is given. Finally, the remaining obstacles and opportunities in the development of vascularized Organ-on-a-Chip platforms for advancing drug development and predictive disease modelling are noted.
Collapse
Affiliation(s)
- Anna Fritschen
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany.
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Germany.
| |
Collapse
|
43
|
Darabi MA, Khosrozadeh A, Wang Y, Ashammakhi N, Alem H, Erdem A, Chang Q, Xu K, Liu Y, Luo G, Khademhosseini A, Xing M. An Alkaline Based Method for Generating Crystalline, Strong, and Shape Memory Polyvinyl Alcohol Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902740. [PMID: 33173720 PMCID: PMC7610272 DOI: 10.1002/advs.201902740] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/28/2020] [Accepted: 02/27/2020] [Indexed: 05/25/2023]
Abstract
Strong, stretchable, and durable biomaterials with shape memory properties can be useful in different biomedical devices, tissue engineering, and soft robotics. However, it is challenging to combine these features. Semi-crystalline polyvinyl alcohol (PVA) has been used to make hydrogels by conventional methods such as freeze-thaw and chemical crosslinking, but it is formidable to produce strong materials with adjustable properties. Herein, a method to induce crystallinity and produce physically crosslinked PVA hydrogels via applying high-concentration sodium hydroxide into dense PVA polymer is introduced. Such a strategy enables the production of physically crosslinked PVA biomaterial with high mechanical properties, low water content, resistance to injury, and shape memory properties. It is also found that the developed PVA hydrogel can recover 90% of plastic deformation due to extension upon supplying water, providing a strong contraction force sufficiently to lift objects 1100 times more than their weight. Cytocompatibility, antifouling property, hemocompatibility, and biocompatibility are also demonstrated in vitro and in vivo. The fabrication methods of PVA-based catheters, injectable electronics, and microfluidic devices are demonstrated. This gelation approach enables both layer-by-layer and 3D printing fabrications.
Collapse
Affiliation(s)
- Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C‐MIT)University of CaliforniaLos AngelesCA90095USA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCA90095USA
- Department of Radiological SciencesDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCA90095USA
- Department of Mechanical EngineeringUniversity of ManitobaWinnipegR3T 5V6Canada
- Terasaki Institute for Biomedical InnovationLos AngelesCA90024USA
| | - Ali Khosrozadeh
- Department of Mechanical EngineeringUniversity of ManitobaWinnipegR3T 5V6Canada
- Department of Physical & Environmental SciencesUniversity of Toronto ScarboroughTorontoOntarioM1C 1A4Canada
| | - Ying Wang
- Institute of Burn ResearchState Key Lab of Trauma Burns and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C‐MIT)University of CaliforniaLos AngelesCA90095USA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCA90095USA
- Department of Radiological SciencesDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCA90095USA
| | - Halima Alem
- Center for Minimally Invasive Therapeutics (C‐MIT)University of CaliforniaLos AngelesCA90095USA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCA90095USA
- Université de LorraineCNRSInstitut Jean Lamour (UMR 7198)Campus Artem 2 allée André Guinier‐BP 50840Nancy CedexF54011France
| | - Ahmet Erdem
- Center for Minimally Invasive Therapeutics (C‐MIT)University of CaliforniaLos AngelesCA90095USA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCA90095USA
- Department of ChemistryKocaeli UniversityUmuttepe CampusKocaeli41380Turkey
- Department of Biomedical EngineeringKocaeli UniversityUmuttepe CampusKocaeli41380Turkey
| | - Qiang Chang
- Department of Mechanical EngineeringUniversity of ManitobaWinnipegR3T 5V6Canada
| | - Kaige Xu
- Department of Mechanical EngineeringUniversity of ManitobaWinnipegR3T 5V6Canada
| | - Yuqing Liu
- Department of Mechanical EngineeringUniversity of ManitobaWinnipegR3T 5V6Canada
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Lab of Trauma Burns and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C‐MIT)University of CaliforniaLos AngelesCA90095USA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCA90095USA
- Department of Radiological SciencesDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCA90095USA
- Terasaki Institute for Biomedical InnovationLos AngelesCA90024USA
- Department of Chemical EngineeringUniversity of CaliforniaLos AngelesCAUSA
| | - Malcolm Xing
- Department of Mechanical EngineeringUniversity of ManitobaWinnipegR3T 5V6Canada
| |
Collapse
|
44
|
Chansoria P, Schuchard K, Shirwaiker RA. Process hybridization schemes for multiscale engineered tissue biofabrication. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1673. [PMID: 33084240 DOI: 10.1002/wnan.1673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Recapitulation of multiscale structure-function properties of cells, cell-secreted extracellular matrix, and 3D architecture of natural tissues is central to engineering biomimetic tissue substitutes. Toward achieving biomimicry, a variety of biofabrication processes have been developed, which can be broadly classified into five categories-fiber and fabric formation, additive manufacturing, surface modification, remote fields, and other notable processes-each with specific advantages and limitations. The majority of biofabrication literature has focused on using a single process at a time, which often limits the range of tissues that could be created with relevant features that span nano to macro scales. With multiscale biomimicry as the goal, development of hybrid biofabrication strategies that synergistically unite two or more processes to complement each other's strengths and limitations has been steadily increasing. This work discusses recent literature in this domain and attempts to equip the reader with the understanding of selecting appropriate processes that can harmonize toward creating engineered tissues with appropriate multiscale structure-function properties. Opportunities related to various hybridization schemes and a future outlook on scale-up biofabrication have also been discussed. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Parth Chansoria
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Karl Schuchard
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Rohan A Shirwaiker
- Edward P. Fitts Department of Industrial and Systems Engineering, North Carolina State University, Raleigh, North Carolina, USA.,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA.,Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, North Carolina, USA
| |
Collapse
|
45
|
Ashammakhi N, Nasiri R, Barros NRD, Tebon P, Thakor J, Goudie M, Shamloo A, Martin MG, Khademhosseini A. Gut-on-a-chip: Current progress and future opportunities. Biomaterials 2020; 255:120196. [PMID: 32623181 PMCID: PMC7396314 DOI: 10.1016/j.biomaterials.2020.120196] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/11/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
Organ-on-a-chip technology tries to mimic the complexity of native tissues in vitro. Important progress has recently been made in using this technology to study the gut with and without microbiota. These in vitro models can serve as an alternative to animal models for studying physiology, pathology, and pharmacology. While these models have greater physiological relevance than two-dimensional (2D) cell systems in vitro, endocrine and immunological functions in gut-on-a-chip models are still poorly represented. Furthermore, the construction of complex models, in which different cell types and structures interact, remains a challenge. Generally, gut-on-a-chip models have the potential to advance our understanding of the basic interactions found within the gut and lay the foundation for future applications in understanding pathophysiology, developing drugs, and personalizing medical treatments.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA.
| | - Rohollah Nasiri
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Natan Roberto de Barros
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA.
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Jai Thakor
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Marcus Goudie
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Martin G Martin
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA.
| |
Collapse
|
46
|
Zhou X, Qu M, Tebon P, Jiang X, Wang C, Xue Y, Zhu J, Zhang S, Oklu R, Sengupta S, Sun W, Khademhosseini A. Screening Cancer Immunotherapy: When Engineering Approaches Meet Artificial Intelligence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001447. [PMID: 33042756 PMCID: PMC7539186 DOI: 10.1002/advs.202001447] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/16/2020] [Indexed: 02/05/2023]
Abstract
Immunotherapy is a class of promising anticancer treatments that has recently gained attention due to surging numbers of FDA approvals and extensive preclinical studies demonstrating efficacy. Nevertheless, further clinical implementation has been limited by high variability in patient response to different immunotherapeutic agents. These treatments currently do not have reliable predictors of efficacy and may lead to side effects. The future development of additional immunotherapy options and the prediction of patient-specific response to treatment require advanced screening platforms associated with accurate and rapid data interpretation. Advanced engineering approaches ranging from sequencing and gene editing, to tumor organoids engineering, bioprinted tissues, and organs-on-a-chip systems facilitate the screening of cancer immunotherapies by recreating the intrinsic and extrinsic features of a tumor and its microenvironment. High-throughput platform development and progress in artificial intelligence can also improve the efficiency and accuracy of screening methods. Here, these engineering approaches in screening cancer immunotherapies are highlighted, and a discussion of the future perspectives and challenges associated with these emerging fields to further advance the clinical use of state-of-the-art cancer immunotherapies are provided.
Collapse
Affiliation(s)
- Xingwu Zhou
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of Chemical and Biomolecular EngineeringHenry Samueli School of Engineering and Applied SciencesUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Moyuan Qu
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Peyton Tebon
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Xing Jiang
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- School of NursingNanjing University of Chinese MedicineNanjing210023China
| | - Canran Wang
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Yumeng Xue
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Jixiang Zhu
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of Biomedical EngineeringSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Shiming Zhang
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Rahmi Oklu
- Minimally Invasive Therapeutics LaboratoryDivision of Vascular and Interventional RadiologyMayo ClinicPhoenixAZ85054USA
| | - Shiladitya Sengupta
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and TechnologyHarvard Medical SchoolBostonMA02115USA
| | - Wujin Sun
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of Chemical and Biomolecular EngineeringHenry Samueli School of Engineering and Applied SciencesUniversity of California, Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of RadiologyDavid Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCA90095USA
- Terasaki Institute for Biomedical InnovationLos AngelesCA90064USA
| |
Collapse
|
47
|
Campbell SB, Wu Q, Yazbeck J, Liu C, Okhovatian S, Radisic M. Beyond Polydimethylsiloxane: Alternative Materials for Fabrication of Organ-on-a-Chip Devices and Microphysiological Systems. ACS Biomater Sci Eng 2020; 7:2880-2899. [PMID: 34275293 DOI: 10.1021/acsbiomaterials.0c00640] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Polydimethylsiloxane (PDMS) is the predominant material used for organ-on-a-chip devices and microphysiological systems (MPSs) due to its ease-of-use, elasticity, optical transparency, and inexpensive microfabrication. However, the absorption of small hydrophobic molecules by PDMS and the limited capacity for high-throughput manufacturing of PDMS-laden devices severely limit the application of these systems in personalized medicine, drug discovery, in vitro pharmacokinetic/pharmacodynamic (PK/PD) modeling, and the investigation of cellular responses to drugs. Consequently, the relatively young field of organ-on-a-chip devices and MPSs is gradually beginning to make the transition to alternative, nonabsorptive materials for these crucial applications. This review examines some of the first steps that have been made in the development of organ-on-a-chip devices and MPSs composed of such alternative materials, including elastomers, hydrogels, thermoplastic polymers, and inorganic materials. It also provides an outlook on where PDMS-alternative devices are trending and the obstacles that must be overcome in the development of versatile devices based on alternative materials to PDMS.
Collapse
Affiliation(s)
- Scott B Campbell
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Qinghua Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Joshua Yazbeck
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Chuan Liu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Sargol Okhovatian
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada.,Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
48
|
Davoodi E, Sarikhani E, Montazerian H, Ahadian S, Costantini M, Swieszkowski W, Willerth S, Walus K, Mofidfar M, Toyserkani E, Khademhosseini A, Ashammakhi N. Extrusion and Microfluidic-based Bioprinting to Fabricate Biomimetic Tissues and Organs. ADVANCED MATERIALS TECHNOLOGIES 2020; 5:1901044. [PMID: 33072855 PMCID: PMC7567134 DOI: 10.1002/admt.201901044] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/10/2020] [Indexed: 05/07/2023]
Abstract
Next generation engineered tissue constructs with complex and ordered architectures aim to better mimic the native tissue structures, largely due to advances in three-dimensional (3D) bioprinting techniques. Extrusion bioprinting has drawn tremendous attention due to its widespread availability, cost-effectiveness, simplicity, and its facile and rapid processing. However, poor printing resolution and low speed have limited its fidelity and clinical implementation. To circumvent the downsides associated with extrusion printing, microfluidic technologies are increasingly being implemented in 3D bioprinting for engineering living constructs. These technologies enable biofabrication of heterogeneous biomimetic structures made of different types of cells, biomaterials, and biomolecules. Microfluiding bioprinting technology enables highly controlled fabrication of 3D constructs in high resolutions and it has been shown to be useful for building tubular structures and vascularized constructs, which may promote the survival and integration of implanted engineered tissues. Although this field is currently in its early development and the number of bioprinted implants is limited, it is envisioned that it will have a major impact on the production of customized clinical-grade tissue constructs. Further studies are, however, needed to fully demonstrate the effectiveness of the technology in the lab and its translation to the clinic.
Collapse
Affiliation(s)
- Elham Davoodi
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Einollah Sarikhani
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Hossein Montazerian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Marco Costantini
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, 00-661 Warsaw, Poland
- Institute of Physical Chemistry – Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Wojciech Swieszkowski
- Biomaterials Group, Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology, 00-661 Warsaw, Poland
| | - Stephanie Willerth
- Department of Mechanical Engineering, Division of Medical Sciences, University of Victoria, BC V8P 5C2, Canada
| | - Konrad Walus
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mohammad Mofidfar
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Ehsan Toyserkani
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
49
|
Pollini M, Paladini F. Bioinspired Materials for Wound Healing Application: The Potential of Silk Fibroin. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E3361. [PMID: 32751205 PMCID: PMC7436046 DOI: 10.3390/ma13153361] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Nature is an incredible source of inspiration for scientific research due to the multiple examples of sophisticated structures and architectures which have evolved for billions of years in different environments. Numerous biomaterials have evolved toward high level functions and performances, which can be exploited for designing novel biomedical devices. Naturally derived biopolymers, in particular, offer a wide range of chances to design appropriate substrates for tissue regeneration and wound healing applications. Wound management still represents a challenging field which requires continuous efforts in scientific research for definition of novel approaches to facilitate and promote wound healing and tissue regeneration, particularly where the conventional therapies fail. Moreover, big concerns associated to the risk of wound infections and antibiotic resistance have stimulated the scientific research toward the definition of products with simultaneous regenerative and antimicrobial properties. Among the bioinspired materials for wound healing, this review focuses attention on a protein derived from the silkworm cocoon, namely silk fibroin, which is characterized by incredible biological features and wound healing capability. As demonstrated by the increasing number of publications, today fibroin has received great attention for providing valuable options for fabrication of biomedical devices and products for tissue engineering. In combination with antimicrobial agents, particularly with silver nanoparticles, fibroin also allows the development of products with improved wound healing and antibacterial properties. This review aims at providing the reader with a comprehensive analysis of the most recent findings on silk fibroin, presenting studies and results demonstrating its effective role in wound healing and its great potential for wound healing applications.
Collapse
Affiliation(s)
- Mauro Pollini
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy
- Caresilk S.r.l.s., Via Monteroni c/o Technological District DHITECH, 73100 Lecce, Italy
| | - Federica Paladini
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy
- Caresilk S.r.l.s., Via Monteroni c/o Technological District DHITECH, 73100 Lecce, Italy
| |
Collapse
|
50
|
Nasiri R, Shamloo A, Akbari J, Tebon P, R. Dokmeci M, Ahadian S. Design and Simulation of an Integrated Centrifugal Microfluidic Device for CTCs Separation and Cell Lysis. MICROMACHINES 2020; 11:E699. [PMID: 32698447 PMCID: PMC7407509 DOI: 10.3390/mi11070699] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 01/02/2023]
Abstract
Separation of circulating tumor cells (CTCs) from blood samples and subsequent DNA extraction from these cells play a crucial role in cancer research and drug discovery. Microfluidics is a versatile technology that has been applied to create niche solutions to biomedical applications, such as cell separation and mixing, droplet generation, bioprinting, and organs on a chip. Centrifugal microfluidic biochips created on compact disks show great potential in processing biological samples for point of care diagnostics. This study investigates the design and numerical simulation of an integrated microfluidic device, including a cell separation unit for isolating CTCs from a blood sample and a micromixer unit for cell lysis on a rotating disk platform. For this purpose, an inertial microfluidic device was designed for the separation of target cells by using contraction-expansion microchannel arrays. Additionally, a micromixer was incorporated to mix separated target cells with the cell lysis chemical reagent to dissolve their membranes to facilitate further assays. Our numerical simulation approach was validated for both cell separation and micromixer units and corroborates existing experimental results. In the first compartment of the proposed device (cell separation unit), several simulations were performed at different angular velocities from 500 rpm to 3000 rpm to find the optimum angular velocity for maximum separation efficiency. By using the proposed inertial separation approach, CTCs, were successfully separated from white blood cells (WBCs) with high efficiency (~90%) at an angular velocity of 2000 rpm. Furthermore, a serpentine channel with rectangular obstacles was designed to achieve a highly efficient micromixer unit with high mixing quality (~98%) for isolated CTCs lysis at 2000 rpm.
Collapse
Affiliation(s)
- Rohollah Nasiri
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran; (R.N.); (J.A.)
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran; (R.N.); (J.A.)
| | - Javad Akbari
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran; (R.N.); (J.A.)
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California—Los Angeles, Los Angeles, CA 90095, USA; (P.T.); (M.R.D.)
- Department of Bioengineering, University of California—Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|