1
|
Amoli MS, Jin L, Rezapourdamanab S, Saadeh M, Singh Y, Ning L, Hwang B, Tomov ML, LaRock CN, Mahmoudi M, Bauser-Heaton H, Serpooshan V. Protocol for 3D bioprinting of nanoparticle-laden hydrogels to enhance antibacterial and imaging properties. STAR Protoc 2024; 5:103393. [PMID: 39412997 PMCID: PMC11530900 DOI: 10.1016/j.xpro.2024.103393] [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: 07/19/2024] [Revised: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
This protocol describes the preparation of a nanoparticle-encapsulated bioink capable of protecting tissue-engineered constructs against bacterial infections while also providing contrast for magnetic resonance (MR) imaging modalities. The report includes details of preparing the methacrylated gelatin-based bioinks and the incorporation of superparamagnetic iron oxide nanoparticles. A detailed protocol is presented for characterizing the bioink, evaluating cell response, and assessing its antibacterial effect. Overall, this article presents a robust approach for the development of antibacterial, MR-visible bioinks suitable for various tissue engineering applications. For complete details on the use and execution of this protocol, please refer to Theus et al.1.
Collapse
Affiliation(s)
- Mehdi Salar Amoli
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA.
| | - Linqi Jin
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Sarah Rezapourdamanab
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Maher Saadeh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Yamini Singh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Mechanical Engineering, Cleveland State University, Cleveland, OH 44115, USA
| | - Boeun Hwang
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Martin L Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher N LaRock
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, GA, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Holly Bauser-Heaton
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Sibley Heart Center at Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| |
Collapse
|
2
|
Hwangbo H, Koo Y, Nacionales F, Kim J, Chae S, Kim GH. Stimulus-assisted in situ bioprinting: advancing direct bench-to-bedside delivery. Trends Biotechnol 2024:S0167-7799(24)00313-5. [PMID: 39643527 DOI: 10.1016/j.tibtech.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 10/04/2024] [Accepted: 11/04/2024] [Indexed: 12/09/2024]
Abstract
The fabrication of 3D bioconstructs using bioprinters will advance the field of regenerative medicine owing to its ability to facilitate clinical treatments. Additional stimulations have been applied to the bioconstructs to guide cells laden in the bioconstructs. However, the conventional bench-to-bedside delivery based on separate bioprinting and biostimulating processes may increase the risks of contamination and shape discordance owing to the considerably long process involved. In situ bioprinting is aimed at eliminating these risks, but stimulation strategies implied during in situ printing have not yet been extensively reviewed. Here, we present the concept of stimulus-assisted in situ bioprinting, which integrates the printing and biostimulation processes by directly applying stimuli to the bioink during fabrication.
Collapse
Affiliation(s)
- Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea; Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - YoungWon Koo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Francis Nacionales
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - JuYeon Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - SooJung Chae
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Geun Hyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea; Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
3
|
You P, Sun H, Chen H, Li C, Mao Y, Zhang T, Yang H, Dong H. Composite bioink incorporating cell-laden liver decellularized extracellular matrix for bioprinting of scaffolds for bone tissue engineering. BIOMATERIALS ADVANCES 2024; 165:214017. [PMID: 39236580 DOI: 10.1016/j.bioadv.2024.214017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
The field of bone tissue engineering (BTE) has witnessed a revolutionary breakthrough with the advent of three-dimensional (3D) bioprinting technology, which is considered an ideal choice for constructing scaffolds for bone regeneration. The key to realizing scaffold biofunctions is the selection and design of an appropriate bioink, and existing bioinks have significant limitations. In this study, a composite bioink based on natural polymers (gelatin and alginate) and liver decellularized extracellular matrix (LdECM) was developed and used to fabricate scaffolds for BTE using 3D bioprinting. Through in vitro studies, the concentration of LdECM incorporated into the bioink was optimized to achieve printability and stability and to improve the proliferation and osteogenic differentiation of loaded rat bone mesenchymal stem cells (rBMSCs). Furthermore, in vivo experiments were conducted using a Sprague Dawley rat model of critical-sized calvarial defects. The proposed rBMSC-laden LdECM-gelatin-alginate scaffold, bioprinted layer-by-layer, was implanted in the rat calvarial defect and the development of new bone growth was studied for four weeks. The findings showed that the proposed bioactive scaffolds facilitated angiogenesis and osteogenesis at the defect site. The findings of this study suggest that the developed rBMSC-laden LdECM-gelatin-alginate bioink has great potential for clinical translation and application in solving bone regeneration problems.
Collapse
Affiliation(s)
- Pengyue You
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Hang Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Haotian Chen
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Changcan Li
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China
| | - Tao Zhang
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China.
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China.
| | - Haitao Dong
- Department of Stomatology, Peking Union Medical College (PUMC) Hospital, Peking Union Medical College (PUMC) & Chinese Academy of Medical Sciences (CAMS), Beijing 100730, China.
| |
Collapse
|
4
|
Masri S, Fauzi MB, Rajab NF, Lee WH, Zainal Abidin DA, Siew EL. In vitro 3D skin culture and its sustainability in toxicology: a narrative review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:476-499. [PMID: 39359233 DOI: 10.1080/21691401.2024.2407617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
In current toxicological research, 2D cell cultures and animal models are well- accepted and commonly employed methods. However, these approaches have many drawbacks and are distant from the actual environment in human. To embrace this, great efforts have been made to provide alternative methods for non-animal skin models in toxicology studies with the need for more mechanistically informative methods. This review focuses on the current state of knowledge regarding the in vitro 3D skin model methods, with different functional states that correspond to the sustainability in the field of toxicology testing. We discuss existing toxicology testing methods using in vitro 3D skin models which provide a better understanding of the testing requirements that are needed. The challenges and future landscape in using the in vitro 3D skin models in toxicology testing are also discussed. We are confident that the in vitro 3D skin models application may become an important tool in toxicology in the context of risk assessment.
Collapse
Affiliation(s)
- Syafira Masri
- Department of Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Nor Fadilah Rajab
- Centre for Health Aging and Wellness, Faculty of Helath Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wing-Hin Lee
- Royal College of Medicine Perak, Universiti Kuala Lumpur (UniKL RCMP), Perak, Malaysia
| | | | - Ee Ling Siew
- ASASIpintar Unit, Pusat PERMATA@Pintar Negara, Universiti Kebangsaan Malaysia, Bangi, Malaysia
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Wu KY, Qian SY, Faucher A, Tran SD. Advancements in Hydrogels for Corneal Healing and Tissue Engineering. Gels 2024; 10:662. [PMID: 39451315 PMCID: PMC11507397 DOI: 10.3390/gels10100662] [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: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant attention for their versatile applications across various fields, including biomedical engineering. This review delves into the fundamentals of hydrogels, exploring their definition, properties, and classification. Hydrogels, as three-dimensional networks of crosslinked polymers, possess tunable properties such as biocompatibility, mechanical strength, and hydrophilicity, making them ideal for medical applications. Uniquely, this article offers original insights into the application of hydrogels specifically for corneal tissue engineering, bridging a gap in current research. The review further examines the anatomical and functional complexities of the cornea, highlighting the challenges associated with corneal pathologies and the current reliance on donor corneas for transplantation. Considering the global shortage of donor corneas, this review discusses the potential of hydrogel-based materials in corneal tissue engineering. Emphasis is placed on the synthesis processes, including physical and chemical crosslinking, and the integration of bioactive molecules. Stimuli-responsive hydrogels, which react to environmental triggers, are identified as promising tools for drug delivery and tissue repair. Additionally, clinical applications of hydrogels in corneal pathologies are explored, showcasing their efficacy in various trials. Finally, the review addresses the challenges of regulatory approval and the need for further research to fully realize the potential of hydrogels in corneal tissue engineering, offering a promising outlook for future developments in this field.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Shu Yu Qian
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Anne Faucher
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
6
|
Francescato R, Moretti M, Bersini S. Endothelial-mesenchymal transition in skeletal muscle: Opportunities and challenges from 3D microphysiological systems. Bioeng Transl Med 2024; 9:e10644. [PMID: 39553431 PMCID: PMC11561840 DOI: 10.1002/btm2.10644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/21/2023] [Accepted: 12/18/2023] [Indexed: 11/19/2024] Open
Abstract
Fibrosis is a pathological condition that in the muscular context is linked to primary diseases such as dystrophies, laminopathies, neuromuscular disorders, and volumetric muscle loss following traumas, accidents, and surgeries. Although some basic mechanisms regarding the role of myofibroblasts in the progression of muscle fibrosis have been discovered, our knowledge of the complex cell-cell, and cell-matrix interactions occurring in the fibrotic microenvironment is still rudimentary. Recently, vascular dysfunction has been emerging as a key hallmark of fibrosis through a process called endothelial-mesenchymal transition (EndoMT). Nevertheless, no effective therapeutic options are currently available for the treatment of muscle fibrosis. This lack is partially due to the absence of advanced in vitro models that can recapitulate the 3D architecture and functionality of a vascularized muscle microenvironment in a human context. These models could be employed for the identification of novel targets and for the screening of potential drugs blocking the progression of the disease. In this review, we explore the potential of 3D human muscle models in studying the role of endothelial cells and EndoMT in muscle fibrotic tissues and identify limitations and opportunities for optimizing the next generation of these microphysiological systems. Starting from the biology of muscle fibrosis and EndoMT, we highlight the synergistic links between different cell populations of the fibrotic microenvironment and how to recapitulate them through microphysiological systems.
Collapse
Affiliation(s)
- Riccardo Francescato
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT)Ente Ospedaliero Cantonale (EOC)BellinzonaSwitzerland
- Service of Orthopaedics and Traumatology, Department of SurgeryEOCLuganoSwitzerland
- Department of ElectronicsInformation and Bioengineering, Politecnico di MilanoMilanoItaly
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT)Ente Ospedaliero Cantonale (EOC)BellinzonaSwitzerland
- Service of Orthopaedics and Traumatology, Department of SurgeryEOCLuganoSwitzerland
- Cell and Tissue Engineering LaboratoryIRCCS Ospedale Galeazzi ‐ Sant'AmbrogioMilanoItaly
- Euler Institute, Faculty of Biomedical SciencesUniversità della Svizzera italiana (USI)LuganoSwitzerland
| | - Simone Bersini
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT)Ente Ospedaliero Cantonale (EOC)BellinzonaSwitzerland
- Service of Orthopaedics and Traumatology, Department of SurgeryEOCLuganoSwitzerland
- Euler Institute, Faculty of Biomedical SciencesUniversità della Svizzera italiana (USI)LuganoSwitzerland
| |
Collapse
|
7
|
Mirsky NA, Ehlen QT, Greenfield JA, Antonietti M, Slavin BV, Nayak VV, Pelaez D, Tse DT, Witek L, Daunert S, Coelho PG. Three-Dimensional Bioprinting: A Comprehensive Review for Applications in Tissue Engineering and Regenerative Medicine. Bioengineering (Basel) 2024; 11:777. [PMID: 39199735 PMCID: PMC11351251 DOI: 10.3390/bioengineering11080777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Since three-dimensional (3D) bioprinting has emerged, it has continuously to evolved as a revolutionary technology in surgery, offering new paradigms for reconstructive and regenerative medical applications. This review highlights the integration of 3D printing, specifically bioprinting, across several surgical disciplines over the last five years. The methods employed encompass a review of recent literature focusing on innovations and applications of 3D-bioprinted tissues and/or organs. The findings reveal significant advances in the creation of complex, customized, multi-tissue constructs that mimic natural tissue characteristics, which are crucial for surgical interventions and patient-specific treatments. Despite the technological advances, the paper introduces and discusses several challenges that remain, such as the vascularization of bioprinted tissues, integration with the host tissue, and the long-term viability of bioprinted organs. The review concludes that while 3D bioprinting holds substantial promise for transforming surgical practices and enhancing patient outcomes, ongoing research, development, and a clear regulatory framework are essential to fully realize potential future clinical applications.
Collapse
Affiliation(s)
| | - Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | - Blaire V. Slavin
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Pelaez
- Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David T. Tse
- Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Paulo G. Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
8
|
Sugiura T, Shahannaz DC, Ferrell BE. Current Status of Cardiac Regenerative Therapy Using Induced Pluripotent Stem Cells. Int J Mol Sci 2024; 25:5772. [PMID: 38891960 PMCID: PMC11171475 DOI: 10.3390/ijms25115772] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Heart failure (HF) is a life-threatening disorder and is treated by drug therapies and surgical interventions such as heart transplantation and left ventricular assist device (LVAD). However, these treatments can lack effectiveness in the long term and are associated with issues such as donor shortage in heart transplantation, and infection, stroke, or gastrointestinal bleeding in LVADs. Therefore, alternative therapeutic strategies are still needed. In this respect, stem cell therapy has been introduced for the treatment of HF and numerous preclinical and clinical studies are employing a range of stem cell varieties. These stem cells, such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have been shown to improve cardiac function and attenuate left ventricular remodeling. IPSCs, which have a capacity for unlimited proliferation and differentiation into cardiomyocytes, are a promising cell source for myocardial regeneration therapy. In this review, we discuss the following topics: (1) what are iPSCs; (2) the limitations and solutions for the translation of iPSC-CMs practically; and (3) the current therapeutic clinical trials.
Collapse
Affiliation(s)
- Tadahisa Sugiura
- Department of Cardiothoracic and Vascular Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, New York, NY 10467, USA; (D.C.S.); (B.E.F.)
| | | | | |
Collapse
|
9
|
Shukla AK, Yoon S, Oh SO, Lee D, Ahn M, Kim BS. Advancement in Cancer Vasculogenesis Modeling through 3D Bioprinting Technology. Biomimetics (Basel) 2024; 9:306. [PMID: 38786516 PMCID: PMC11118135 DOI: 10.3390/biomimetics9050306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Cancer vasculogenesis is a pivotal focus of cancer research and treatment given its critical role in tumor development, metastasis, and the formation of vasculogenic microenvironments. Traditional approaches to investigating cancer vasculogenesis face significant challenges in accurately modeling intricate microenvironments. Recent advancements in three-dimensional (3D) bioprinting technology present promising solutions to these challenges. This review provides an overview of cancer vasculogenesis and underscores the importance of precise modeling. It juxtaposes traditional techniques with 3D bioprinting technologies, elucidating the advantages of the latter in developing cancer vasculogenesis models. Furthermore, it explores applications in pathological investigations, preclinical medication screening for personalized treatment and cancer diagnostics, and envisages future prospects for 3D bioprinted cancer vasculogenesis models. Despite notable advancements, current 3D bioprinting techniques for cancer vasculogenesis modeling have several limitations. Nonetheless, by overcoming these challenges and with technological advances, 3D bioprinting exhibits immense potential for revolutionizing the understanding of cancer vasculogenesis and augmenting treatment modalities.
Collapse
Affiliation(s)
- Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sik Yoon
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea
| | - Sae-Ock Oh
- Research Center for Molecular Control of Cancer Cell Diversity, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea
| | - Minjun Ahn
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
10
|
Albrecht FB, Ahlfeld T, Klatt A, Heine S, Gelinsky M, Kluger PJ. Biofabrication's Contribution to the Evolution of Cultured Meat. Adv Healthc Mater 2024; 13:e2304058. [PMID: 38339837 PMCID: PMC11468272 DOI: 10.1002/adhm.202304058] [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: 01/25/2024] [Indexed: 02/12/2024]
Abstract
Cultured Meat (CM) is a growing field in cellular agriculture, driven by the environmental impact of conventional meat production, which contributes to climate change and occupies ≈70% of arable land. As demand for meat alternatives rises, research in this area expands. CM production relies on tissue engineering techniques, where a limited number of animal cells are cultured in vitro and processed to create meat-like tissue comprising muscle and adipose components. Currently, CM is primarily produced on a small scale in pilot facilities. Producing a large cell mass based on suitable cell sources and bioreactors remains challenging. Advanced manufacturing methods and innovative materials are required to subsequently process this cell mass into CM products on a large scale. Consequently, CM is closely linked with biofabrication, a suite of technologies for precisely arranging cellular aggregates and cell-material composites to construct specific structures, often using robotics. This review provides insights into contemporary biomedical biofabrication technologies, focusing on significant advancements in muscle and adipose tissue biofabrication for CM production. Novel materials for biofabricating CM are also discussed, emphasizing their edibility and incorporation of healthful components. Finally, initial studies on biofabricated CM are examined, addressing current limitations and future challenges for large-scale production.
Collapse
Affiliation(s)
| | - Tilman Ahlfeld
- Technische Universität DresdenCentre for Translational BoneJoint and Soft Tissue Research01307DresdenGermany
| | - Annemarie Klatt
- Reutlingen UniversityReutlingen Research Institute72762ReutlingenGermany
| | - Simon Heine
- Reutlingen UniversityReutlingen Research Institute72762ReutlingenGermany
| | - Michael Gelinsky
- Technische Universität DresdenCentre for Translational BoneJoint and Soft Tissue Research01307DresdenGermany
| | | |
Collapse
|
11
|
Zhu L, Cheng C, Liu S, Yang L, Han P, Cui T, Zhang Y. Advancements and application prospects of three-dimensional models for primary liver cancer: a comprehensive review. Front Bioeng Biotechnol 2023; 11:1343177. [PMID: 38188493 PMCID: PMC10771299 DOI: 10.3389/fbioe.2023.1343177] [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: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Primary liver cancer (PLC) is one of the most commonly diagnosed cancers worldwide and a leading cause of cancer-related deaths. However, traditional liver cancer models fail to replicate tumor heterogeneity and the tumor microenvironment, limiting the study and personalized treatment of liver cancer. To overcome these limitations, scientists have introduced three-dimensional (3D) culture models as an emerging research tool. These 3D models, utilizing biofabrication technologies such as 3D bioprinting and microfluidics, enable more accurate simulation of the in vivo tumor microenvironment, replicating cell morphology, tissue stiffness, and cell-cell interactions. Compared to traditional two-dimensional (2D) models, 3D culture models better mimic tumor heterogeneity, revealing differential sensitivity of tumor cell subpopulations to targeted therapies or immunotherapies. Additionally, these models can be used to assess the efficacy of potential treatments, providing guidance for personalized therapy. 3D liver cancer models hold significant value in tumor biology, understanding the mechanisms of disease progression, and drug screening. Researchers can gain deeper insights into the impact of the tumor microenvironment on tumor cells and their interactions with the surrounding milieu. Furthermore, these models allow for the evaluation of treatment responses, offering more accurate guidance for clinical interventions. In summary, 3D models provide a realistic and reliable tool for advancing PLC research. By simulating tumor heterogeneity and the microenvironment, these models contribute to a better understanding of the disease mechanisms and offer new strategies for personalized treatment. Therefore, 3D models hold promising prospects for future PLC research.
Collapse
Affiliation(s)
- Liuyang Zhu
- First Central Clinical College of Tianjin Medical University, Tianjin, China
| | | | - Sen Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Long Yang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Pinsheng Han
- Nankai University of Medicine College, Tianjin, China
| | - Tao Cui
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, China
- Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Beijing, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
12
|
Abadpour S, Niemi EM, Orrhult LS, Hermanns C, de Vries R, Nogueira LP, Haugen HJ, Josefsen D, Krauss S, Gatenholm P, van Apeldoorn A, Scholz H. Adipose-Derived Stromal Cells Preserve Pancreatic Islet Function in a Transplantable 3D Bioprinted Scaffold. Adv Healthc Mater 2023; 12:e2300640. [PMID: 37781993 PMCID: PMC11469278 DOI: 10.1002/adhm.202300640] [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: 02/27/2023] [Revised: 09/07/2023] [Indexed: 10/03/2023]
Abstract
Intra-portal islet transplantation is currently the only clinically approved beta cell replacement therapy, but its outcome is hindered by limited cell survival due to a multifactorial reaction against the allogeneic tissue in liver. Adipose-derived stromal cells (ASCs) can potentially improve the islet micro-environment by their immunomodulatory action. The challenge is to combine both islets and ASCs in a relatively easy and consistent long-term manner in a deliverable scaffold. Manufacturing the 3D bioprinted double-layered scaffolds with primary islets and ASCs using a mix of alginate/nanofibrillated cellulose (NFC) bioink is reported. The diffusion properties of the bioink and the supportive effect of human ASCs on islet viability, glucose sensing, insulin secretion, and reducing the secretion of pro-inflammatory cytokines are demonstrated. Diabetic mice transplanted with islet-ASC scaffolds reach normoglycemia seven days post-transplantation with no significant difference between this group and the group received islets under the kidney capsules. In addition, animals transplanted with islet-ASC scaffolds stay normoglycemic and show elevated levels of C-peptide compared to mice transplanted with islet-only scaffolds. The data present a functional 3D bioprinted scaffold for islets and ASCs transplanted to the extrahepatic site and suggest a possible role of ASCs on improving the islet micro-environment.
Collapse
Affiliation(s)
- Shadab Abadpour
- Department of Transplant MedicineOslo University HospitalOslo0372Norway
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
| | - Essi M. Niemi
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Department of Vascular SurgeryAker HospitalOslo University HospitalOslo0586Norway
| | - Linnea Strid Orrhult
- 3D Bioprinting CenterWWSCDepartment of Chemistry and Chemical EngineeringChalmers University of TechnologyGothenburg41296Sweden
| | - Carolin Hermanns
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | - Rick de Vries
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | | | | | - Dag Josefsen
- Section for Cellular TherapyRadiumhospitaletOslo University HospitalOslo0379Norway
| | - Stefan Krauss
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Department of Immunology and Transfusion MedicineOslo University HospitalOslo0372Norway
| | - Paul Gatenholm
- 3D Bioprinting CenterWWSCDepartment of Chemistry and Chemical EngineeringChalmers University of TechnologyGothenburg41296Sweden
- CELLHEAL ASSandvika1337Norway
| | - Aart van Apeldoorn
- MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229The Netherlands
| | - Hanne Scholz
- Department of Transplant MedicineOslo University HospitalOslo0372Norway
- Institute for Surgical ResearchOslo University HospitalOslo0372Norway
- Hybrid Technology Hub – Centre of ExcellenceInstitute of Basic Medical SciencesUniversity of OsloOslo0372Norway
- Section for Cellular TherapyRadiumhospitaletOslo University HospitalOslo0379Norway
| |
Collapse
|
13
|
Chi M, Yuan B, Xie Z, Hong J. The Innovative Biomaterials and Technologies for Developing Corneal Endothelium Tissue Engineering Scaffolds: A Review and Prospect. Bioengineering (Basel) 2023; 10:1284. [PMID: 38002407 PMCID: PMC10669703 DOI: 10.3390/bioengineering10111284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Corneal transplantation is the only treatment for corneal endothelial blindness. However, there is an urgent need to find substitutes for corneal endothelium grafts due to the global shortage of donor corneas. An emerging research field focuses on the construction of scaffold-based corneal endothelium tissue engineering (CETE). Long-term success in CETE transplantation may be achieved by selecting the appropriate biomaterials as scaffolds of corneal endothelial cells and adding bioactive materials to promote cell activity. This article reviews the research progress of CETE biomaterials in the past 20 years, describes the key characteristics required for corneal endothelial scaffolds, and summarizes the types of materials that have been reported. Based on these, we list feasible improvement strategies for biomaterials innovation. In addition, we describe the improved techniques for the scaffolds' surface topography and drug delivery system. Some promising technologies for constructing CETE are proposed. However, some questions have not been answered yet, and clinical trials and industrialization should be carried out with caution.
Collapse
Affiliation(s)
- Miaomiao Chi
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China; (M.C.); (B.Y.); (Z.X.)
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Bowei Yuan
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China; (M.C.); (B.Y.); (Z.X.)
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Zijun Xie
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China; (M.C.); (B.Y.); (Z.X.)
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Jing Hong
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China; (M.C.); (B.Y.); (Z.X.)
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| |
Collapse
|
14
|
Deptuła M, Zawrzykraj M, Sawicka J, Banach-Kopeć A, Tylingo R, Pikuła M. Application of 3D- printed hydrogels in wound healing and regenerative medicine. Biomed Pharmacother 2023; 167:115416. [PMID: 37683592 DOI: 10.1016/j.biopha.2023.115416] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Hydrogels are three-dimensional polymer networks with hydrophilic properties. The modifiable properties of hydrogels and the structure resembling living tissue allow their versatile application. Therefore, increasing attention is focused on the use of hydrogels as bioinks for three-dimensional (3D) printing in tissue engineering. Bioprinting involves the fabrication of complex structures from several types of materials, cells, and bioactive compounds. Stem cells (SC), such as mesenchymal stromal cells (MSCs) are frequently employed in 3D constructs. SCs have desirable biological properties such as the ability to differentiate into various types of tissue and high proliferative capacity. Encapsulating SCs in 3D hydrogel constructs enhances their reparative abilities and improves the likelihood of reaching target tissues. In addition, created constructs can simulate the tissue environment and mimic biological signals. Importantly, the immunogenicity of scaffolds is minimized through the use of patient-specific cells and the biocompatibility and biodegradability of the employed biopolymers. Regenerative medicine is taking advantage of the aforementioned capabilities in regenerating various tissues- muscle, bones, nerves, heart, skin, and cartilage.
Collapse
Affiliation(s)
- Milena Deptuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Medical University of Gdansk, Poland.
| | | | - Justyna Sawicka
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Poland
| | - Adrianna Banach-Kopeć
- Department of Chemistry, Technology and Biochemistry of Food, Faculty of Chemistry, Gdansk University of Technology, Poland
| | - Robert Tylingo
- Department of Chemistry, Technology and Biochemistry of Food, Faculty of Chemistry, Gdansk University of Technology, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Medical University of Gdansk, Poland
| |
Collapse
|
15
|
Sánchez-Duffhues G, Hiepen C. Human iPSCs as Model Systems for BMP-Related Rare Diseases. Cells 2023; 12:2200. [PMID: 37681932 PMCID: PMC10487005 DOI: 10.3390/cells12172200] [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: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Disturbances in bone morphogenetic protein (BMP) signalling contribute to onset and development of a number of rare genetic diseases, including Fibrodysplasia ossificans progressiva (FOP), Pulmonary arterial hypertension (PAH), and Hereditary haemorrhagic telangiectasia (HHT). After decades of animal research to build a solid foundation in understanding the underlying molecular mechanisms, the progressive implementation of iPSC-based patient-derived models will improve drug development by addressing drug efficacy, specificity, and toxicity in a complex humanized environment. We will review the current state of literature on iPSC-derived model systems in this field, with special emphasis on the access to patient source material and the complications that may come with it. Given the essential role of BMPs during embryonic development and stem cell differentiation, gain- or loss-of-function mutations in the BMP signalling pathway may compromise iPSC generation, maintenance, and differentiation procedures. This review highlights the need for careful optimization of the protocols used. Finally, we will discuss recent developments towards complex in vitro culture models aiming to resemble specific tissue microenvironments with multi-faceted cellular inputs, such as cell mechanics and ECM together with organoids, organ-on-chip, and microfluidic technologies.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), ISPA-HUCA, Avda. de Roma, s/n, 33011 Oviedo, Spain
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Christian Hiepen
- Department of Engineering and Natural Sciences, Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665 Recklinghausen, Germany
| |
Collapse
|
16
|
Cardinale V, Lanthier N, Baptista PM, Carpino G, Carnevale G, Orlando G, Angelico R, Manzia TM, Schuppan D, Pinzani M, Alvaro D, Ciccocioppo R, Uygun BE. Cell transplantation-based regenerative medicine in liver diseases. Stem Cell Reports 2023; 18:1555-1572. [PMID: 37557073 PMCID: PMC10444572 DOI: 10.1016/j.stemcr.2023.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 08/11/2023] Open
Abstract
This review aims to evaluate the current preclinical state of liver bioengineering, the clinical context for liver cell therapies, the cell sources, the delivery routes, and the results of clinical trials for end-stage liver disease. Different clinical settings, such as inborn errors of metabolism, acute liver failure, chronic liver disease, liver cirrhosis, and acute-on-chronic liver failure, as well as multiple cellular sources were analyzed; namely, hepatocytes, hepatic progenitor cells, biliary tree stem/progenitor cells, mesenchymal stromal cells, and macrophages. The highly heterogeneous clinical scenario of liver disease and the availability of multiple cellular sources endowed with different biological properties make this a multidisciplinary translational research challenge. Data on each individual liver disease and more accurate endpoints are urgently needed, together with a characterization of the regenerative pathways leading to potential therapeutic benefit. Here, we critically review these topics and identify related research needs and perspectives in preclinical and clinical settings.
Collapse
Affiliation(s)
- Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Nicolas Lanthier
- Service d'Hépato-gastroentérologie, Cliniques Universitaires Saint-Luc, Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd), Madrid, Spain; Fundación ARAID, Zaragoza, Spain; Department of Biomedical and Aerospace Engineering, Universidad Carlos III de Madrid, Madrid, Spain
| | - Guido Carpino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry, and Morphological Sciences with Interest in Transplant, Oncology, and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giuseppe Orlando
- Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Roberta Angelico
- Hepatobiliary Surgery and Transplant Unit, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tommaso Maria Manzia
- Hepatobiliary Surgery and Transplant Unit, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Detlef Schuppan
- Institute of Translational Immunology, Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
| | - Domenico Alvaro
- Department of Translation and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy.
| | - Basak E Uygun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
17
|
Juraski AC, Sharma S, Sparanese S, da Silva VA, Wong J, Laksman Z, Flannigan R, Rohani L, Willerth SM. 3D bioprinting for organ and organoid models and disease modeling. Expert Opin Drug Discov 2023; 18:1043-1059. [PMID: 37431937 DOI: 10.1080/17460441.2023.2234280] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION 3D printing, a versatile additive manufacturing technique, has diverse applications ranging from transportation, rapid prototyping, clean energy, and medical devices. AREAS COVERED The authors focus on how 3D printing technology can enhance the drug discovery process through automating tissue production that enables high-throughput screening of potential drug candidates. They also discuss how the 3D bioprinting process works and what considerations to address when using this technology to generate cell laden constructs for drug screening as well as the outputs from such assays necessary for determining the efficacy of potential drug candidates. They focus on how bioprinting how has been used to generate cardiac, neural, and testis tissue models, focusing on bio-printed 3D organoids. EXPERT OPINION The next generation of 3D bioprinted organ model holds great promises for the field of medicine. In terms of drug discovery, the incorporation of smart cell culture systems and biosensors into 3D bioprinted models could provide highly detailed and functional organ models for drug screening. By addressing current challenges of vascularization, electrophysiological control, and scalability, researchers can obtain more reliable and accurate data for drug development, reducing the risk of drug failures during clinical trials.
Collapse
Affiliation(s)
- Amanda C Juraski
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Department of Chemical Engineering, Polytechnic School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sonali Sharma
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sydney Sparanese
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Victor A da Silva
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
| | - Julie Wong
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Zachary Laksman
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Leili Rohani
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| |
Collapse
|
18
|
Antonelli F. 3D Cell Models in Radiobiology: Improving the Predictive Value of In Vitro Research. Int J Mol Sci 2023; 24:10620. [PMID: 37445795 DOI: 10.3390/ijms241310620] [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: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer is intrinsically complex, comprising both heterogeneous cellular composition and extracellular matrix. In vitro cancer research models have been widely used in the past to model and study cancer. Although two-dimensional (2D) cell culture models have traditionally been used for cancer research, they have many limitations, such as the disturbance of interactions between cellular and extracellular environments and changes in cell morphology, polarity, division mechanism, differentiation and cell motion. Moreover, 2D cell models are usually monotypic. This implies that 2D tumor models are ineffective at accurately recapitulating complex aspects of tumor cell growth, as well as their radiation responses. Over the past decade there has been significant uptake of three-dimensional (3D) in vitro models by cancer researchers, highlighting a complementary model for studies of radiation effects on tumors, especially in conjunction with chemotherapy. The introduction of 3D cell culture approaches aims to model in vivo tissue interactions with radiation by positioning itself halfway between 2D cell and animal models, and thus opening up new possibilities in the study of radiation response mechanisms of healthy and tumor tissues.
Collapse
Affiliation(s)
- Francesca Antonelli
- Laboratory of Biomedical Technologies, Division of Health Protection Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00123 Rome, Italy
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Bioengineering of functional cardiac tissue composed of primary cardiomyocytes has great potential for myocardial regeneration and in vitro tissue modeling. 3D bioprinting was developed to create cardiac tissue in hydrogels that can mimic the structural, physiological, and functional features of native myocardium. Through a detailed review of the 3D printing technologies and bioink materials used in the creation of a heart tissue, this article discusses the potential of engineered heart tissues in biomedical applications. RECENT FINDINGS In this review, we discussed the recent progress in 3D bioprinting strategies for cardiac tissue engineering, including bioink and 3D bioprinting methods as well as examples of engineered cardiac tissue such as in vitro cardiac models and vascular channels. 3D printing is a powerful tool for creating in vitro cardiac tissues that are structurally and functionally similar to real tissues. The use of human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) enables the generation of patient-specific tissues. These tissues have the potential to be used for regenerative therapies, disease modeling, and drug testing.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Materials Science and Engineering Program, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Yi Xiang
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Min Tang
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Shaochen Chen
- Materials Science and Engineering Program, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
- Department of Bioengineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| |
Collapse
|
20
|
Nguyen HT, Peirsman A, Tirpakova Z, Mandal K, Vanlauwe F, Maity S, Kawakita S, Khorsandi D, Herculano R, Umemura C, Yilgor C, Bell R, Hanson A, Li S, Nanda HS, Zhu Y, Najafabadi AH, Jucaud V, Barros N, Dokmeci MR, Khademhosseini A. Engineered Vasculature for Cancer Research and Regenerative Medicine. MICROMACHINES 2023; 14:978. [PMID: 37241602 PMCID: PMC10221678 DOI: 10.3390/mi14050978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
Engineered human tissues created by three-dimensional cell culture of human cells in a hydrogel are becoming emerging model systems for cancer drug discovery and regenerative medicine. Complex functional engineered tissues can also assist in the regeneration, repair, or replacement of human tissues. However, one of the main hurdles for tissue engineering, three-dimensional cell culture, and regenerative medicine is the capability of delivering nutrients and oxygen to cells through the vasculatures. Several studies have investigated different strategies to create a functional vascular system in engineered tissues and organ-on-a-chips. Engineered vasculatures have been used for the studies of angiogenesis, vasculogenesis, as well as drug and cell transports across the endothelium. Moreover, vascular engineering allows the creation of large functional vascular conduits for regenerative medicine purposes. However, there are still many challenges in the creation of vascularized tissue constructs and their biological applications. This review will summarize the latest efforts to create vasculatures and vascularized tissues for cancer research and regenerative medicine.
Collapse
Affiliation(s)
- Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Zuzana Tirpakova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Florian Vanlauwe
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Rondinelli Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Bioengineering & Biomaterials Group, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil
| | - Christian Umemura
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Adrian Hanson
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Himansu Sekhar Nanda
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Biomedical Engineering and Technology Laboratory, PDPM—Indian Institute of Information Technology Design Manufacturing, Jabalpur 482005, Madhya Pradesh, India
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Natan Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| |
Collapse
|
21
|
Flores-Torres S, Jiang T, Kort-Mascort J, Yang Y, Peza-Chavez O, Pal S, Mainolfi A, Pardo LA, Ferri L, Bertos N, Sangwan V, Kinsella JM. Constructing 3D In Vitro Models of Heterocellular Solid Tumors and Stromal Tissues Using Extrusion-Based Bioprinting. ACS Biomater Sci Eng 2023; 9:542-561. [PMID: 36598339 DOI: 10.1021/acsbiomaterials.2c00998] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Malignant tumor tissues exhibit inter- and intratumoral heterogeneities, aberrant development, dynamic stromal composition, diverse tissue phenotypes, and cell populations growing within localized mechanical stresses in hypoxic conditions. Experimental tumor models employing engineered systems that isolate and study these complex variables using in vitro techniques are under development as complementary methods to preclinical in vivo models. Here, advances in extrusion bioprinting as an enabling technology to recreate the three-dimensional tumor milieu and its complex heterogeneous characteristics are reviewed. Extrusion bioprinting allows for the deposition of multiple materials, or selected cell types and concentrations, into models based upon physiological features of the tumor. This affords the creation of complex samples with representative extracellular or stromal compositions that replicate the biology of patient tissue. Biomaterial engineering of printable materials that replicate specific features of the tumor microenvironment offer experimental reproducibility, throughput, and physiological relevance compared to animal models. In this review, we describe the potential of extrusion-based bioprinting to recreate the tumor microenvironment within in vitro models.
Collapse
Affiliation(s)
| | - Tao Jiang
- Department of Intelligent Machinery and Instrument, College of Intelligence Science and Technology, National University of Defense Technology Changsha, Hunan 410073, China
| | | | - Yun Yang
- Department of Intelligent Machinery and Instrument, College of Intelligence Science and Technology, National University of Defense Technology Changsha, Hunan 410073, China
| | - Omar Peza-Chavez
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Sanjima Pal
- Department of Surgery, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Alisia Mainolfi
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Lucas Antonio Pardo
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Lorenzo Ferri
- Department of Surgery, McGill University, Montreal, Quebec H3G 2M1, Canada.,Department of Medicine, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Nicholas Bertos
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec H4A 3J1, Canada
| | - Veena Sangwan
- Department of Surgery, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Joseph M Kinsella
- Department of Bioengineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| |
Collapse
|
22
|
Cadamuro F, Nicotra F, Russo L. 3D printed tissue models: From hydrogels to biomedical applications. J Control Release 2023; 354:726-745. [PMID: 36682728 DOI: 10.1016/j.jconrel.2023.01.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
The development of new advanced constructs resembling structural and functional properties of human organs and tissues requires a deep knowledge of the morphological and biochemical properties of the extracellular matrices (ECM), and the capacity to reproduce them. Manufacturing technologies like 3D printing and bioprinting represent valuable tools for this purpose. This review will describe how morphological and biochemical properties of ECM change in different tissues, organs, healthy and pathological states, and how ECM mimics with the required properties can be generated by 3D printing and bioprinting. The review describes and classifies the polymeric materials of natural and synthetic origin exploited to generate the hydrogels acting as "inks" in the 3D printing process, with particular emphasis on their functionalization allowing crosslinking and conjugation with signaling molecules to develop bio-responsive and bio-instructive ECM mimics.
Collapse
Affiliation(s)
- Francesca Cadamuro
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Francesco Nicotra
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Laura Russo
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY Galway, Ireland.
| |
Collapse
|
23
|
Jia S, Bu Y, Lau DSA, Lin Z, Sun T, Lu WW, Lu S, Ruan C, Chan CHJ. Advances in 3D bioprinting technology for functional corneal reconstruction and regeneration. Front Bioeng Biotechnol 2023; 10:1065460. [PMID: 36686254 PMCID: PMC9852906 DOI: 10.3389/fbioe.2022.1065460] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Corneal transplantation constitutes one of the major treatments in severe cases of corneal diseases. The lack of cornea donors as well as other limitations of corneal transplantation necessitate the development of artificial corneal substitutes. Biosynthetic cornea model using 3D printing technique is promising to generate artificial corneal structure that can resemble the structure of the native human cornea and is applicable for regenerative medicine. Research on bioprinting artificial cornea has raised interest into the wide range of materials and cells that can be utilized as bioinks for optimal clarity, biocompatibility, and tectonic strength. With continued advances in biomaterials science and printing technology, it is believed that bioprinted cornea will eventually achieve a level of clinical functionality and practicality as to replace donated corneal tissues, with their associated limitations such as limited or unsteady supply, and possible infectious disease transmission. Here, we review the literature on bioprinting strategies, 3D corneal modelling, material options, and cellularization strategies in relation to keratoprosthesis design. The progress, limitations and expectations of recent cases of 3D bioprinting of artifial cornea are discussed. An outlook on the rise of 3D bioprinting in corneal reconstruction and regeneration is provided.
Collapse
Affiliation(s)
- Shuo Jia
- Department of Ophthalmology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yashan Bu
- Department of Ophthalmology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Dzi-Shing Aaron Lau
- Department of Orthopedic and Traumatology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhizhen Lin
- Department of Ophthalmology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Tianhao Sun
- Department of Orthopedic and Traumatology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Gangqing Biomedical Technology Co. Ltd, Shenzhen, China
| | - Weijia William Lu
- Department of Orthopedic and Traumatology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sheng Lu
- Department of Orthopedic Surgery, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Changshun Ruan
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheuk-Hung Jonathan Chan
- Department of Ophthalmology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
24
|
Yang P, Ju Y, Hu Y, Xie X, Fang B, Lei L. Emerging 3D bioprinting applications in plastic surgery. Biomater Res 2023; 27:1. [PMID: 36597149 PMCID: PMC9808966 DOI: 10.1186/s40824-022-00338-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Plastic surgery is a discipline that uses surgical methods or tissue transplantation to repair, reconstruct and beautify the defects and deformities of human tissues and organs. Three-dimensional (3D) bioprinting has gained widespread attention because it enables fine customization of the implants in the patient's surgical area preoperatively while avoiding some of the adverse reactions and complications of traditional surgical approaches. In this paper, we review the recent research advances in the application of 3D bioprinting in plastic surgery. We first introduce the printing process and basic principles of 3D bioprinting technology, revealing the advantages and disadvantages of different bioprinting technologies. Then, we describe the currently available bioprinting materials, and dissect the rationale for special dynamic 3D bioprinting (4D bioprinting) that is achieved by varying the combination strategy of bioprinting materials. Later, we focus on the viable clinical applications and effects of 3D bioprinting in plastic surgery. Finally, we summarize and discuss the challenges and prospects for the application of 3D bioprinting in plastic surgery. We believe that this review can contribute to further development of 3D bioprinting in plastic surgery and provide lessons for related research.
Collapse
Affiliation(s)
- Pu Yang
- grid.452708.c0000 0004 1803 0208Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 People’s Republic of China
| | - Yikun Ju
- grid.452708.c0000 0004 1803 0208Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 People’s Republic of China
| | - Yue Hu
- grid.449525.b0000 0004 1798 4472School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000 People’s Republic of China
| | - Xiaoyan Xie
- grid.452708.c0000 0004 1803 0208Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011 People’s Republic of China
| | - Bairong Fang
- grid.452708.c0000 0004 1803 0208Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 People’s Republic of China
| | - Lanjie Lei
- grid.263826.b0000 0004 1761 0489School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 People’s Republic of China
| |
Collapse
|
25
|
Guagliano G, Volpini C, Briatico-Vangosa F, Cornaglia AI, Visai L, Petrini P. Toward 3D-Bioprinted Models of the Liver to Boost Drug Development. Macromol Biosci 2022; 22:e2200264. [PMID: 36106413 DOI: 10.1002/mabi.202200264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/06/2022] [Indexed: 01/15/2023]
Abstract
The main problems in drug development are connected to enormous costs related to the paltry success rate. The current situation empowered the development of high-throughput and reliable instruments, in addition to the current golden standards, able to predict the failures in the early preclinical phase. Being hepatotoxicity responsible for the failure of 30% of clinical trials, and the 21% of withdrawal of marketed drugs, the development of complex in vitro models (CIVMs) of liver is currently one of the hottest topics in the field. Among the different fabrication techniques, 3D-bioprinting is emerging as a powerful ally for their production, allowing the manufacture of three-dimensional constructs characterized by computer-controlled and customized geometry, and inter-batches reproducibility. Thanks to these, it is possible to rapidly produce tailored cell-laden constructs, to be cultured within static and dynamic systems, thus reaching a further degree of personalization when designing in vitro models. This review highlights and prioritizes the most recent advances related to the development of CIVMs of the hepatic environment to be specifically applied to pharmaceutical research, with a special focus on 3D-bioprinting, since the liver is primarily involved in the metabolism of drugs.
Collapse
Affiliation(s)
- Giuseppe Guagliano
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, Milano, MI, 20133, Italy
| | - Cristina Volpini
- Molecular Medicine Department (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Via Forlanini 14, Pavia, PV, 27100, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio Di Nanotecnologie, ICS Maugeri IRCCS, Via S. Boezio 28, Pavia, PV, 27100, Italy
| | - Francesco Briatico-Vangosa
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, Milano, MI, 20133, Italy
| | - Antonia Icaro Cornaglia
- University of Pavia - Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit, Via Forlanini 2, Pavia, PV, 27100, Italy
| | - Livia Visai
- Molecular Medicine Department (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Via Forlanini 14, Pavia, PV, 27100, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio Di Nanotecnologie, ICS Maugeri IRCCS, Via S. Boezio 28, Pavia, PV, 27100, Italy.,Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), Università di Pavia Unit, Pavia, PV, 27100, Italy
| | - Paola Petrini
- Department of Chemistry, Materials, and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, Milano, MI, 20133, Italy.,Interuniversity Center for the promotion of the 3Rs principles in teaching and research (Centro 3R), Politecnico di Milano Unit, Milano, MI, 20133, Italy
| |
Collapse
|
26
|
Dornhof J, Zieger V, Kieninger J, Frejek D, Zengerle R, Urban GA, Kartmann S, Weltin A. Bioprinting-based automated deposition of single cancer cell spheroids into oxygen sensor microelectrode wells. LAB ON A CHIP 2022; 22:4369-4381. [PMID: 36254669 DOI: 10.1039/d2lc00705c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Three-dimensional (3D) cell agglomerates, such as microtissues, organoids, and spheroids, become increasingly relevant in biomedicine. They can provide in vitro models that recapitulate functions of the original tissue in the body and have applications in cancer research. For example, they are widely used in organ-on-chip systems. Microsensors can provide essential real-time information on cell metabolism as well as the reliability and quality of culture conditions. The combination of sensors and 3D cell cultures, especially single spheroids, is challenging in terms of reproducible formation, manipulation, and access to spheroids, precise positioning near sensors, and high cell-to-volume ratios to obtain meaningful biosignals in the most parallel approach possible. To overcome this challenge, we combined state-of-the-art bioprinting techniques to automatically print tumour spheroids directly into microwells of a chip-based electrochemical oxygen sensor array. We demonstrated highly accurate and reproducible spheroid formation (diameter of approx. 200 μm) and a spheroid deposition precision of 25 μm within a volume of 22 nl per droplet. Microstructures and hydrogel-coated microwells allowed the placement of single MCF-7 breast cancer spheroids close to the sensor electrodes. The microelectrode wells were sealed for oxygen measurements within a 55 nl volume for fast concentration changes. Accurate and stable amperometric oxygen sensor performance was demonstrated from atmospheric to anoxic regions. Cellular respiration rates from single tumour spheroids in the range of 450-850 fmol min-1 were determined, and alterations of cell metabolism upon drug exposure were shown. Our results uniquely combine bioprinting with 3D cell culture monitoring and demonstrate the much-needed effort for facilitation, parallelization, sensor integration, and drug delivery in 3D cell culture and organ-on-chip experiments. The workflow has a high degree of automation and potential for scalability. In order to achieve greater flexibility in the automation of spheroid formation and trapping, we employed a method based on drop-on-demand liquid handling systems, instead of the typical on-chip approach commonly used in microfluidics. Its relevance ranges from fundamental metabolic research over standardization of cell culture experiments and toxicological studies, to personalized medicine, e.g. patient-specific chemotherapy.
Collapse
Affiliation(s)
- Johannes Dornhof
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Viktoria Zieger
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Jochen Kieninger
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | | | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
- Hahn-Schickard, Freiburg, Germany
| | - Gerald A Urban
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | - Sabrina Kartmann
- Laboratory for MEMS Applications, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
- Hahn-Schickard, Freiburg, Germany
| | - Andreas Weltin
- Laboratory for Sensors, IMTEK - Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
27
|
Can 3D bioprinting solve the mystery of senescence in cancer therapy? Ageing Res Rev 2022; 81:101732. [PMID: 36100069 DOI: 10.1016/j.arr.2022.101732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/30/2022] [Accepted: 09/08/2022] [Indexed: 01/31/2023]
Abstract
Tumor dormancy leading to cancer relapse is still a poorly understood mechanism. Several cell states such as quiescence and diapause can explain the persistence of tumor cells in a dormant state, but the potential role of tumor cell senescence has been met with hesitance given the historical understanding of the senescent growth arrest as irreversible. However, recent evidence has suggested that senescence might contribute to dormancy and relapse, although its exact role is not fully developed. This limited understanding is largely due to the paucity of reliable study models. The current 2D cell modeling is overly simplistic and lacks the appropriate representation of the interactions between tumor cells (senescent or non-senescent) and the other cell types within the tumor microenvironment (TME), as well as with the extracellular matrix (ECM). 3D cell culture models, including 3D bioprinting techniques, offer a promising approach to better recapitulate the native cancer microenvironment and would significantly improve our understanding of cancer biology and cellular response to treatment, particularly Therapy-Induced Senescence (TIS), and its contribution to tumor dormancy and cancer recurrence. Fabricating a novel 3D bioprinted model offers excellent opportunities to investigate both the role of TIS in tumor dormancy and the utility of senolytics (drugs that selectively eliminate senescent cells) in targeting dormant cancer cells and mitigating the risk for resurgence. In this review, we discuss literature on the possible contribution of TIS in tumor dormancy, provide examples on the current 3D models of senescence, and propose a novel 3D model to investigate the ultimate role of TIS in mediating overall response to therapy.
Collapse
|
28
|
Groth T, Stegmayr BG, Ash SR, Kuchinka J, Wieringa FP, Fissell WH, Roy S. Wearable and implantable artificial kidney devices for end-stage kidney disease treatment-Current status and review. Artif Organs 2022; 47:649-666. [PMID: 36129158 DOI: 10.1111/aor.14396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a major cause of early death worldwide. By 2030, 14.5 million people will have end-stage kidney disease (ESKD, or CKD stage 5), yet only 5.4 million will receive kidney replacement therapy (KRT) due to economic, social, and political factors. Even for those who are offered KRT by various means of dialysis, the life expectancy remains far too low. OBSERVATION Researchers from different fields of artificial organs collaborate to overcome the challenges of creating products such as Wearable and/or Implantable Artificial Kidneys capable of providing long-term effective physiologic kidney functions such as removal of uremic toxins, electrolyte homeostasis, and fluid regulation. A focus should be to develop easily accessible, safe, and inexpensive KRT options that enable a good quality of life and will also be available for patients in less-developed regions of the world. CONCLUSIONS Hence, it is required to discuss some of the limits and burdens of transplantation and different techniques of dialysis, including those performed at home. Furthermore, hurdles must be considered and overcome to develop wearable and implantable artificial kidney devices that can help to improve the quality of life and life expectancy of patients with CKD.
Collapse
Affiliation(s)
- Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,International Federation for Artificial Organs, Painesville, Ohio, USA
| | - Bernd G Stegmayr
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| | | | - Janna Kuchinka
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Fokko P Wieringa
- IMEC, Eindhoven, The Netherlands.,Department of Nephrology, University Medical Centre, Utrecht, The Netherlands.,European Kidney Health Alliance, WG3 "Breakthrough Innovation", Brussels, Belgium
| | | | - Shuvo Roy
- University of California, California, San Francisco, USA
| |
Collapse
|
29
|
Mohr E, Thum T, Bär C. Accelerating Cardiovascular Research: Recent Advances in Translational 2D and 3D Heart Models. Eur J Heart Fail 2022; 24:1778-1791. [PMID: 35867781 DOI: 10.1002/ejhf.2631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/30/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022] Open
Abstract
In vitro modelling the complex (patho-) physiological conditions of the heart is a major challenge in cardiovascular research. In recent years, methods based on three-dimensional (3D) cultivation approaches have steadily evolved to overcome the major limitations of conventional adherent monolayer cultivation (2D). These 3D approaches aim to study, reproduce or modify fundamental native features of the heart such as tissue organization and cardiovascular microenvironment. Therefore, these systems have great potential for (patient-specific) disease research, for the development of new drug screening platforms, and for the use in regenerative and replacement therapy applications. Consequently, continuous improvement and adaptation is required with respect to fundamental limitations such as cardiomyocyte maturation, scalability, heterogeneity, vascularization, and reproduction of native properties. In this review, 2D monolayer culturing and the 3D in vitro systems of cardiac spheroids, organoids, engineered cardiac microtissue and bioprinting as well as the ex vivo technique of myocardial slicing are introduced with their basic concepts, advantages, and limitations. Furthermore, recent advances of various new approaches aiming to extend as well as to optimize these in vitro and ex vivo systems are presented. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Elisa Mohr
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
30
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|