1
|
Zhou R, Brislinger D, Fuchs J, Lyons A, Langthaler S, Hauser CAE, Baumgartner C. Vascularised organoids: Recent advances and applications in cancer research. Clin Transl Med 2025; 15:e70258. [PMID: 40045486 PMCID: PMC11882480 DOI: 10.1002/ctm2.70258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
Organoids are three-dimensional (3D) cellular models designed to replicate human tissues and organs while preserving their physiological complexity and functionality. Among these, vascularised organoids represent a groundbreaking advancement in 3D tissue engineering, incorporating vascular networks into engineered tissues to more accurately mimic the in vivo tumour microenvironment. These models offer significantly improved physiological relevance compared to conventional two-dimensional cultures or animal models, positioning them as invaluable tools in cancer research. Despite their potential, the rapid proliferation of techniques and materials for developing vascularised organoids presents challenges for researchers navigating this dynamic field. This systematic review provides a comprehensive examination of methodologies for fabricating vascularised organoids, with a focus on strategies that enhance vascularisation and support organoid growth. It critically evaluates the materials used, emphasising those that effectively mimic the extracellular matrix and facilitate vascular network formation. Key advancements in engineered organoids models are highlighted, emphasising their potential for studying interactions between vasculature and cancer cells, conducting drug screening, and understanding cytokine regulation. In summary, this review provides an in-depth overview of the current landscape of vascularised organoid fabrication and functionality, addressing challenges and opportunities within the field. A detailed understanding of the scope and future trajectories is essential for advancing organoid development and expanding their applications in both basic cancer research and clinical practice. KEY POINTS: Comparative analysis: Evaluation of organoids, animal models, and 2D models, highlighting their respective strengths and limitations in replicating physiological conditions and studying disease processes. Vascularisation techniques: Comparative evaluation of vascularised organoid fabrication methods, emphasising their efficiency, scalability and ability to replicate physiological vascular networks. Material selection: Thorough evaluation of materials for vascularised organoid culture system, focusing on those that effectively mimic the extracellular matrix and support vascular network formation. Applications: Overview of organoid applications in basic cancer research and clinical settings, with an emphasis on their potential in drug discovery, disease modelling and exploring complex biological processes.
Collapse
Affiliation(s)
- Rui Zhou
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Dagmar Brislinger
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Julia Fuchs
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Alicia Lyons
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Sonja Langthaler
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Charlotte A. E. Hauser
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Christian Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| |
Collapse
|
2
|
Tripathi S, Dash M, Chakraborty R, Lukman HJ, Kumar P, Hassan S, Mehboob H, Singh H, Nanda HS. Engineering considerations in the design of tissue specific bioink for 3D bioprinting applications. Biomater Sci 2024; 13:93-129. [PMID: 39535021 DOI: 10.1039/d4bm01192a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Over eight million surgical procedures are conducted annually in the United Stats to address organ failure or tissue losses. In response to this pressing need, recent medical advancements have significantly improved patient outcomes, primarily through innovative reconstructive surgeries utilizing tissue grafting techniques. Despite tremendous efforts, repairing damaged tissues remains a major clinical challenge for bioengineers and clinicians. 3D bioprinting is an additive manufacturing technique that holds significant promise for creating intricately detailed constructs of tissues, thereby bridging the gap between engineered and actual tissue constructs. In contrast to non-biological printing, 3D bioprinting introduces added intricacies, including considerations for material selection, cell types, growth, and differentiation factors. However, technical challenges arise, particularly concerning the delicate nature of living cells in bioink for tissue construction and limited knowledge about the cell fate processes in such a complex biomechanical environment. A bioink must have appropriate viscoelastic and rheological properties to mimic the native tissue microenvironment and attain desired biomechanical properties. Hence, the properties of bioink play a vital role in the success of 3D bioprinted substitutes. This review comprehensively delves into the scientific aspects of tissue-centric or tissue-specific bioinks and sheds light on the current challenges of the translation of bioinks and bioprinting.
Collapse
Affiliation(s)
- Shivi Tripathi
- Biomaterials and Biomanufacturing Laboratory, Discipline of Mechanical Engineering, PDPM Indian Institute of Information Technology Design and Manufacturing, Jabalpur 482005, MP, India.
- International Centre for Sustainable and Net Zero Technologies, PDPM-Indian Institute of Information Technology Design and Manufacturing Jabalpur, Madhya Pradesh 482005, India
| | - Madhusmita Dash
- School of Minerals, Metallurgical and Materials Engineering, Indian Institute of Technology Bhubaneswar, Argul, Khordha, Odisha 752050, India
| | - Ruchira Chakraborty
- Biodesign and Medical Device Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Harri Junaedi Lukman
- Department of Engineering and Management, College of Engineering, Prince Sultan University, Riyadh 12435, Saudi Arabia
| | - Prasoon Kumar
- Biodesign and Medical Device Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, 769008, Odisha, India.
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
- Biotechnology Centre (BTC), Khalifa University, Abu Dhabi, United Arab Emirates
| | - Hassan Mehboob
- Department of Engineering and Management, College of Engineering, Prince Sultan University, Riyadh 12435, Saudi Arabia
| | - Harpreet Singh
- Dr B R Ambedkar National Institute of Technology Jalandhar, Grand Trunk Road, Barnala Amritsar Bypass Rd, Jalandhar, Punjab 14401111, India
| | - Himansu Sekhar Nanda
- Biomaterials and Biomanufacturing Laboratory, Discipline of Mechanical Engineering, PDPM Indian Institute of Information Technology Design and Manufacturing, Jabalpur 482005, MP, India.
- International Centre for Sustainable and Net Zero Technologies, PDPM-Indian Institute of Information Technology Design and Manufacturing Jabalpur, Madhya Pradesh 482005, India
- Terasaki Institute for Biomedical Innovation, 21100 Erwin, St Los Angeles, CA 91367, USA
| |
Collapse
|
3
|
Gokaltun A, Asik E, Byrne D, Yarmush ML, Usta OB. Supercooled preservation of cultured primary rat hepatocyte monolayers. Front Bioeng Biotechnol 2024; 12:1429412. [PMID: 39076209 PMCID: PMC11284110 DOI: 10.3389/fbioe.2024.1429412] [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: 05/08/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Supercooled preservation (SCP) is a technology that involves cooling a substance below its freezing point without initiating ice crystal formation. It is a promising alternative to prolong the preservation time of cells, tissues, engineered tissue products, and organs compared to the current practices of hypothermic storage. Two-dimensional (2D) engineered tissues are extensively used in in vitro research for drug screening and development and investigation of disease progression. Despite their widespread application, there is a lack of research on the SCP of 2D-engineered tissues. In this study, we presented the effects of SCP at -2 and -6°C on primary rat hepatocyte (PRH) monolayers for the first time and compared cell viability and functionality with cold storage (CS, + 4°C). We preserved PRH monolayers in two different commercially available solutions: Hypothermosol-FRS (HTS-FRS) and the University of Wisconsin (UW) with and without supplements (i.e., polyethylene glycol (PEG) and 3-O-Methyl-Α-D-Glucopyranose (3-OMG)). Our findings revealed that UW with and without supplements were inadequate for the short-term preservation of PRH monolayers for both SCP and CS with high viability, functionality, and monolayer integrity. The combination of supplements (PEG and 3-OMG) in the HTS-FRS solution outperformed the other groups and yielded the highest viability and functional capacity. Notably, PRH monolayers exhibited superior viability and functionality when stored at -2°C through SCP for up to 3 days compared to CS. Overall, our results demonstrated that SCP is a feasible approach to improving the short-term preservation of PRH monolayers and enables readily available 2D-engineered tissues to advance in vitro research. Furthermore, our findings provide insights into preservation outcomes across various biological levels, from cells to tissues and organs, contributing to the advancement of bioengineering and biotechnology.
Collapse
Affiliation(s)
- Aslihan Gokaltun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, United States
- Department of Chemical Engineering, Hacettepe University, Ankara, Türkiye
| | - Eda Asik
- Shriners Hospitals for Children, Boston, MA, United States
- Department of Bioengineering, Hacettepe University, Ankara, Türkiye
| | - Delaney Byrne
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
| | - Martin L. Yarmush
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
- Department of Biomedical Engineering, Rutgers University, Newark, NJ, United States
| | - O. Berk Usta
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
| |
Collapse
|
4
|
Albrecht FB, Schmidt FF, Schmidt C, Börret R, Kluger PJ. Robot-based 6D bioprinting for soft tissue biomedical applications. Eng Life Sci 2024; 24:e2300226. [PMID: 38975018 PMCID: PMC11223372 DOI: 10.1002/elsc.202300226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 03/08/2024] [Accepted: 05/10/2024] [Indexed: 07/09/2024] Open
Abstract
Within this interdisciplinary study, we demonstrate the applicability of a 6D printer for soft tissue engineering models. For this purpose, a special plant was constructed, combining the technical requirements for 6D printing with the biological necessities, especially for soft tissue. Therefore, a commercial 6D robot arm was combined with a sterilizable housing (including a high-efficiency particulate air (HEPA) filter and ultraviolet radiation (UVC) lamps) and a custom-made printhead and printbed. Both components allow cooling and heating, which is desirable for working with viable cells. In addition, a spraying unit was installed that allows the distribution of fine droplets of a liquid. Advanced geometries on uneven or angled surfaces can be created with the use of all six axes. Based on often used bioinks in the field of soft tissue engineering (gellan gum, collagen, and gelatin methacryloyl) with very different material properties, we could demonstrate the flexibility of the printing system. Furthermore, cell-containing constructs using primary human adipose-derived stem cells (ASCs) could be produced in an automated manner. In addition to cell survival, the ability to differentiate along the adipogenic lineage could also be demonstrated as a representative of soft tissue engineering.
Collapse
Affiliation(s)
- Franziska B. Albrecht
- Reutlingen Research InstituteReutlingen UniversityReutlingenGermany
- Faculty of Natural ScienceUniversity of HohenheimStuttgartGermany
| | - Freia F. Schmidt
- Reutlingen Research InstituteReutlingen UniversityReutlingenGermany
| | | | - Rainer Börret
- Aalen University, Center for Optical TechnologiesAalenGermany
| | - Petra J. Kluger
- Faculty of Life SciencesReutlingen UniversityReutlingenGermany
| |
Collapse
|
5
|
De Giorgio G, Matera B, Vurro D, Manfredi E, Galstyan V, Tarabella G, Ghezzi B, D'Angelo P. Silk Fibroin Materials: Biomedical Applications and Perspectives. Bioengineering (Basel) 2024; 11:167. [PMID: 38391652 PMCID: PMC10886036 DOI: 10.3390/bioengineering11020167] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/13/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
The golden rule in tissue engineering is the creation of a synthetic device that simulates the native tissue, thus leading to the proper restoration of its anatomical and functional integrity, avoiding the limitations related to approaches based on autografts and allografts. The emergence of synthetic biocompatible materials has led to the production of innovative scaffolds that, if combined with cells and/or bioactive molecules, can improve tissue regeneration. In the last decade, silk fibroin (SF) has gained attention as a promising biomaterial in regenerative medicine due to its enhanced bio/cytocompatibility, chemical stability, and mechanical properties. Moreover, the possibility to produce advanced medical tools such as films, fibers, hydrogels, 3D porous scaffolds, non-woven scaffolds, particles or composite materials from a raw aqueous solution emphasizes the versatility of SF. Such devices are capable of meeting the most diverse tissue needs; hence, they represent an innovative clinical solution for the treatment of bone/cartilage, the cardiovascular system, neural, skin, and pancreatic tissue regeneration, as well as for many other biomedical applications. The present narrative review encompasses topics such as (i) the most interesting features of SF-based biomaterials, bare SF's biological nature and structural features, and comprehending the related chemo-physical properties and techniques used to produce the desired formulations of SF; (ii) the different applications of SF-based biomaterials and their related composite structures, discussing their biocompatibility and effectiveness in the medical field. Particularly, applications in regenerative medicine are also analyzed herein to highlight the different therapeutic strategies applied to various body sectors.
Collapse
Affiliation(s)
- Giuseppe De Giorgio
- IMEM-CNR, Institute of Materials for Electronics and Magnetism-National Research Council, Parco Area delle Scienze 37/A, 43124 Parma, Italy
| | - Biagio Matera
- Center of Dental Medicine, Department of Medicine and Surgery, University of Parma, Via Gramsci 14/A, 43126 Parma, Italy
| | - Davide Vurro
- IMEM-CNR, Institute of Materials for Electronics and Magnetism-National Research Council, Parco Area delle Scienze 37/A, 43124 Parma, Italy
| | - Edoardo Manfredi
- Center of Dental Medicine, Department of Medicine and Surgery, University of Parma, Via Gramsci 14/A, 43126 Parma, Italy
| | - Vardan Galstyan
- IMEM-CNR, Institute of Materials for Electronics and Magnetism-National Research Council, Parco Area delle Scienze 37/A, 43124 Parma, Italy
- Department of Engineering "Enzo Ferrari", University of Modena and Reggio Emilia, Via Vivarelli 10, 41125 Modena, Italy
| | - Giuseppe Tarabella
- IMEM-CNR, Institute of Materials for Electronics and Magnetism-National Research Council, Parco Area delle Scienze 37/A, 43124 Parma, Italy
| | - Benedetta Ghezzi
- IMEM-CNR, Institute of Materials for Electronics and Magnetism-National Research Council, Parco Area delle Scienze 37/A, 43124 Parma, Italy
- Center of Dental Medicine, Department of Medicine and Surgery, University of Parma, Via Gramsci 14/A, 43126 Parma, Italy
| | - Pasquale D'Angelo
- IMEM-CNR, Institute of Materials for Electronics and Magnetism-National Research Council, Parco Area delle Scienze 37/A, 43124 Parma, Italy
| |
Collapse
|
6
|
De Francesco F, Zingaretti N, Parodi PC, Riccio M. The Evolution of Current Concept of the Reconstructive Ladder in Plastic Surgery: The Emerging Role of Translational Medicine. Cells 2023; 12:2567. [PMID: 37947645 PMCID: PMC10649097 DOI: 10.3390/cells12212567] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Plastic surgeons have used the reconstructive ladder for many decades as a standard directory for complex trauma reconstruction with the goal of repairing body structures and restoring functionality. This consists of different surgical maneuvers, such as secondary intention and direct tissue closure, as well as more complex methods such as local tissue transfer and free flap. The reconstructive ladder represents widely known options achievable for tissue reconstruction and wound closure that puts at the bottom rung the simplest methods of reconstruction and strengthens the complexity by moving upward. Regenerative medicine and surgery constitute a quickly spreading area of translational research that can be employed by minimally invasive surgical strategies, with the aim of regenerating cells and tissues in vivo in order to reestablish normal function through the intrinsic potential of cells, in combination with biomaterials and appropriate biochemical stimuli. These translational procedures have the aim of creating an appropriate microenvironment capable of supporting the physiological cellular function to generate the desired cells or tissues and to generate parenchymal, stromal, and vascular components on demand, and above all to produce intelligent materials capable of determining the fate of cells. Smart technologies have been grown that give extra "rungs" on the classic reconstructive ladder to integrate a more holistic, patient-based approach with improved outcomes. This commentary presents the evolution of the traditional concept of the reconstructive ladder in the field of plastic surgery into a new course with the aim of achieving excellent results for soft tissue reconstruction by applying innovative technologies and biologically active molecules for a wide range of surgical diseases.
Collapse
Affiliation(s)
- Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy;
| | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Pier Camillo Parodi
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, 33100 Udine, Italy; (N.Z.); (P.C.P.)
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, University Hospital (AOU Ospedali Riuniti di Ancona), Via Conca 71, Torrette di Ancona, 60123 Ancona, Italy;
| |
Collapse
|
7
|
Stossi F, Singh PK, Safari K, Marini M, Labate D, Mancini MA. High throughput microscopy and single cell phenotypic image-based analysis in toxicology and drug discovery. Biochem Pharmacol 2023; 216:115770. [PMID: 37660829 DOI: 10.1016/j.bcp.2023.115770] [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: 06/01/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Measuring single cell responses to the universe of chemicals (drugs, natural products, environmental toxicants etc.) is of paramount importance to human health as phenotypic variability in sensing stimuli is a hallmark of biology that is considered during high throughput screening. One of the ways to approach this problem is via high throughput, microscopy-based assays coupled with multi-dimensional single cell analysis methods. Here, we will summarize some of the efforts in this vast and growing field, focusing on phenotypic screens (e.g., Cell Painting), single cell analytics and quality control, with particular attention to environmental toxicology and drug screening. We will discuss advantages and limitations of high throughput assays with various end points and levels of complexity.
Collapse
Affiliation(s)
- Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA.
| | - Pankaj K Singh
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Kazem Safari
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Michela Marini
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Demetrio Labate
- GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Department of Mathematics, University of Houston, Houston, TX, USA
| | - Michael A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; GCC Center for Advanced Microscopy and Image Informatics, Houston, TX, USA; Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
8
|
Jeyachandran D, Murshed M, Haglund L, Cerruti M. A Bioglass-Poly(lactic-co-glycolic Acid) Scaffold@Fibrin Hydrogel Construct to Support Endochondral Bone Formation. Adv Healthc Mater 2023; 12:e2300211. [PMID: 37462089 PMCID: PMC11468889 DOI: 10.1002/adhm.202300211] [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: 01/19/2023] [Revised: 06/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Bone tissue engineering using stem cells to build bone directly on a scaffold matrix often fails due to lack of oxygen at the injury site. This may be avoided by following the endochondral ossification route; herein, a cartilage template is promoted first, which can survive hypoxic environments, followed by its hypertrophy and ossification. However, hypertrophy is so far only achieved using biological factors. This work introduces a Bioglass-Poly(lactic-co-glycolic acid@fibrin (Bg-PLGA@fibrin) construct where a fibrin hydrogel infiltrates and encapsulates a porous Bg-PLGA. The hypothesis is that mesenchymal stem cells (MSCs) loaded in the fibrin gel and induced into chondrogenesis degrade the gel and become hypertrophic upon reaching the stiffer, bioactive Bg-PLGA core, without external induction factors. Results show that Bg-PLGA@fibrin induces hypertrophy, as well as matrix mineralization and osteogenesis; it also promotes a change in morphology of the MSCs at the gel/scaffold interface, possibly a sign of osteoblast-like differentiation of hypertrophic chondrocytes. Thus, the Bg-PLGA@fibrin construct can sequentially support the different phases of endochondral ossification purely based on material cues. This may facilitate clinical translation by decreasing in-vitro cell culture time pre-implantation and the complexity associated with the use of external induction factors.
Collapse
Affiliation(s)
| | - Monzur Murshed
- Faculty of DentistryDepartment of Medicineand Shriners Hospital for ChildrenMcGill UniversityMontrealQuebecH4A 0A9Canada
| | - Lisbet Haglund
- Experimental SurgeryMcGill UniversityMontrealH3G 2M1Canada
| | - Marta Cerruti
- Department of Mining and Materials EngineeringMcGill UniversityMontrealH3A 0C1Canada
| |
Collapse
|
9
|
Donkers JM, van der Vaart JI, van de Steeg E. Gut-on-a-Chip Research for Drug Development: Implications of Chip Design on Preclinical Oral Bioavailability or Intestinal Disease Studies. Biomimetics (Basel) 2023; 8:226. [PMID: 37366821 PMCID: PMC10296225 DOI: 10.3390/biomimetics8020226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
The gut plays a key role in drug absorption and metabolism of orally ingested drugs. Additionally, the characterization of intestinal disease processes is increasingly gaining more attention, as gut health is an important contributor to our overall health. The most recent innovation to study intestinal processes in vitro is the development of gut-on-a-chip (GOC) systems. Compared to conventional in vitro models, they offer more translational value, and many different GOC models have been presented over the past years. Herein, we reflect on the almost unlimited choices in designing and selecting a GOC for preclinical drug (or food) development research. Four components that largely influence the GOC design are highlighted, namely (1) the biological research questions, (2) chip fabrication and materials, (3) tissue engineering, and (4) the environmental and biochemical cues to add or measure in the GOC. Examples of GOC studies in the two major areas of preclinical intestinal research are presented: (1) intestinal absorption and metabolism to study the oral bioavailability of compounds, and (2) treatment-orientated research for intestinal diseases. The last section of this review presents an outlook on the limitations to overcome in order to accelerate preclinical GOC research.
Collapse
Affiliation(s)
- Joanne M. Donkers
- Department of Metabolic Health Research, TNO, Sylviusweg 71, 2333 BE Leiden, The Netherlands; (J.I.v.d.V.); (E.v.d.S.)
| | - Jamie I. van der Vaart
- Department of Metabolic Health Research, TNO, Sylviusweg 71, 2333 BE Leiden, The Netherlands; (J.I.v.d.V.); (E.v.d.S.)
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Evita van de Steeg
- Department of Metabolic Health Research, TNO, Sylviusweg 71, 2333 BE Leiden, The Netherlands; (J.I.v.d.V.); (E.v.d.S.)
| |
Collapse
|
10
|
Valls-Esteve A, Lustig-Gainza P, Adell-Gomez N, Tejo-Otero A, Englí-Rueda M, Julian-Alvarez E, Navarro-Sureda O, Fenollosa-Artés F, Rubio-Palau J, Krauel L, Munuera J. A state-of-the-art guide about the effects of sterilization processes on 3D-printed materials for surgical planning and medical applications: A comparative study. Int J Bioprint 2023; 9:756. [PMID: 37555083 PMCID: PMC10406103 DOI: 10.18063/ijb.756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/01/2023] [Indexed: 08/10/2023] Open
Abstract
Surgeons use different medical devices in the surgery, such as patient-specific anatomical models, cutting and positioning guides, or implants. These devices must be sterilized before being used in the operation room. There are many sterilization processes available, with autoclave, hydrogen peroxide, and ethylene oxide being the most common in hospital settings. Each method has both advantages and disadvantages in terms of mechanics, chemical interaction, and post-treatment accuracy. The aim of the present study is to evaluate the dimensional and mechanical effect of the most commonly used sterilization techniques available in clinical settings, i.e., Autoclave 121, Autoclave 134, and hydrogen peroxide (HPO), on 11 of the most used 3D-printed materials fabricated using additive manufacturing technologies. The results showed that the temperature (depending on the sterilization method) and the exposure time to that temperature influence not only the mechanical behavior but also the original dimensioning planned on the 3D model. Therefore, HPO is a better overall option for most of the materials evaluated. Finally, based on the results of the study, a recommendation guide on sterilization methods per material, technology, and clinical application is presented.
Collapse
Affiliation(s)
- Arnau Valls-Esteve
- Innovation Department, Hospital Sant Joan de Déu,
Esplugues de Llobregat, Spain
- Medicina i Recerca Translacional, Facultat de Medicina i
Ciències de la Salut, Universitat de Barcelona, Spain
- 3D for Health Unit (3D4H), Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
| | | | - Nuria Adell-Gomez
- Innovation Department, Hospital Sant Joan de Déu,
Esplugues de Llobregat, Spain
- 3D for Health Unit (3D4H), Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
| | - Aitor Tejo-Otero
- Centre CIM, Universitat Politècnica de Catalunya
(CIM UPC), Barcelona, Spain
| | - Marti Englí-Rueda
- Innovation Department, Hospital Sant Joan de Déu,
Esplugues de Llobregat, Spain
- 3D for Health Unit (3D4H), Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
| | | | - Osmeli Navarro-Sureda
- Sterilization Department, Hospital Sant Joan de Déu,
Universitat de Barcelona, Spain
| | - Felip Fenollosa-Artés
- Centre CIM, Universitat Politècnica de Catalunya
(CIM UPC), Barcelona, Spain
- Department of Mechanical Engineering, School of Engineering
of Barcelona (ETSEIB), Universitat Politècnica de Catalunya, Barcelona,
Spain
| | - Josep Rubio-Palau
- Medicina i Recerca Translacional, Facultat de Medicina i
Ciències de la Salut, Universitat de Barcelona, Spain
- 3D for Health Unit (3D4H), Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
- Department of Pediatric Surgery, Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
- Maxillofacial Unit, Department of Pediatric Surgery,
Hospital Sant Joan de Déu, Universitat de Barcelona, Spain
| | - Lucas Krauel
- Medicina i Recerca Translacional, Facultat de Medicina i
Ciències de la Salut, Universitat de Barcelona, Spain
- 3D for Health Unit (3D4H), Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
- Department of Pediatric Surgery, Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
| | - Josep Munuera
- Medicina i Recerca Translacional, Facultat de Medicina i
Ciències de la Salut, Universitat de Barcelona, Spain
- 3D for Health Unit (3D4H), Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
- Department of Diagnostic Imaging, Hospital Sant Joan de
Déu, Universitat de Barcelona, Spain
| |
Collapse
|
11
|
Manduca N, Maccafeo E, De Maria R, Sistigu A, Musella M. 3D cancer models: One step closer to in vitro human studies. Front Immunol 2023; 14:1175503. [PMID: 37114038 PMCID: PMC10126361 DOI: 10.3389/fimmu.2023.1175503] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer immunotherapy is the great breakthrough in cancer treatment as it displayed prolonged progression-free survival over conventional therapies, yet, to date, in only a minority of patients. In order to broad cancer immunotherapy clinical applicability some roadblocks need to be overcome, first among all the lack of preclinical models that faithfully depict the local tumor microenvironment (TME), which is known to dramatically affect disease onset, progression and response to therapy. In this review, we provide the reader with a detailed overview of current 3D models developed to mimick the complexity and the dynamics of the TME, with a focus on understanding why the TME is a major target in anticancer therapy. We highlight the advantages and translational potentials of tumor spheroids, organoids and immune Tumor-on-a-Chip models in disease modeling and therapeutic response, while outlining pending challenges and limitations. Thinking forward, we focus on the possibility to integrate the know-hows of micro-engineers, cancer immunologists, pharmaceutical researchers and bioinformaticians to meet the needs of cancer researchers and clinicians interested in using these platforms with high fidelity for patient-tailored disease modeling and drug discovery.
Collapse
Affiliation(s)
- Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ‘A. Gemelli’ - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
12
|
Pansai N, Detarun P, Chinnaworn A, Sangsupawanich P, Wichienchot S. Effects of dragon fruit oligosaccharides on immunity, gut microbiome, and their metabolites in healthy adults – a randomized double-blind placebo controlled study. Food Res Int 2023; 167:112657. [PMID: 37087207 DOI: 10.1016/j.foodres.2023.112657] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/26/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
Healthy food has wide popularity and relates positively to health. Our previous studies have shown that dragon fruit oligosaccharides (DFO) have prebiotic activities, balancing the gut microbiota in a simulated human colon system, and are safe and stimulate the immune system in rats. The effects of DFO on immune stimulation gut microbe modulation and the correlation of gut microbiota and nutrients were investigated in a human trial. This clinical study was a randomized, double-blinded, placebo-controlled trial. The participants were 107 healthy adults, divided into 3 groups that received DFO in drinking waterdoses of 4 and 8 g/day, compared to the placebo group for 4 consecutive weeks. DFO consumption at 4 g/day increased IgA level (11.31 mg/dL or 10.95% from baseline) and 8 g/day outstandingly promoted the growth of Bifidobacterium spp. (8.41%) and Faecalibacterium (1.99%) and decreased harmful bacteria, especially, Escherichia coli (8.44%). The relationship between gut microbes and nutrient intake was explored and significant (p < 0.05) correlations between specific microbial groups and intakes of specific macro- and micronutrients were observed. The potential dose of DFO for healthy adults was established as 4 g/day for improving IgA level and 8 g/day for promoting beneficial gut microbiota.
Collapse
|
13
|
Dozzo A, Galvin A, Shin JW, Scalia S, O'Driscoll CM, Ryan KB. Modelling acute myeloid leukemia (AML): What's new? A transition from the classical to the modern. Drug Deliv Transl Res 2022:10.1007/s13346-022-01189-4. [PMID: 35930221 DOI: 10.1007/s13346-022-01189-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/24/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous malignancy affecting myeloid cells in the bone marrow (BM) but can spread giving rise to impaired hematopoiesis. AML incidence increases with age and is associated with poor prognostic outcomes. There has been a disconnect between the success of novel drug compounds observed in preclinical studies of hematological malignancy and less than exceptional therapeutic responses in clinical trials. This review aims to provide a state-of-the-art overview on the different preclinical models of AML available to expand insights into disease pathology and as preclinical screening tools. Deciphering the complex physiological and pathological processes and developing predictive preclinical models are key to understanding disease progression and fundamental in the development and testing of new effective drug treatments. Standard scaffold-free suspension models fail to recapitulate the complex environment where AML occurs. To this end, we review advances in scaffold/matrix-based 3D models and outline the most recent advances in on-chip technology. We also provide an overview of clinically relevant animal models and review the expanding use of patient-derived samples, which offer the prospect to create more "patient specific" screening tools either in the guise of 3D matrix models, microphysiological "organ-on-chip" tools or xenograft models and discuss representative examples.
Collapse
Affiliation(s)
| | - Aoife Galvin
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, 909 S. Wolcott Ave, Chicago, IL, 5091 COMRB, USA
| | - Santo Scalia
- Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Caitriona M O'Driscoll
- School of Pharmacy, University College Cork, Cork, Ireland.,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland. .,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
14
|
Co-Culture of THP-1 Cells and Normal Human Epidermal Keratinocytes (NHEK) for Modified Human Cell Line Activation Test (h-CLAT). APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12126207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
To improve the accuracy of skin sensitization prediction of chemicals by conventional alternative methods using cells, it is important to reproduce the environment of skin in vitro, such as the crosstalk between keratinocytes and dendritic cells (DCs). We developed a skin sensitization test system based on the markers and criteria of the human cell line activation test (h-CLAT), which combines THP-1 cells as a surrogate for DCs and keratinized normal human epidermal keratinocytes (NHEK). After exposure to chemicals via keratinized NHEK, the cell surface expression of CD54 and CD86 on THP-1 was measured by flow cytometry. This co-culture system evaluated 2,4-dinitrochlorobenzene (DNCB), a typical sensitizer, as positive, lactic acid (LA), a non-sensitizer, as negative, and isoeugenol (IE), a prohapten that requires biological activation to acquire skin sensitization, as positive. However, the expression levels of CD54 and CD86 in DNCB-treated THP-1 were lower than those in normal h-CLAT. Therefore, we investigated the effects of the medium and secretion by NHEK cells on THP-1 cells. CD54 and CD86 expression was enhanced in monocultured THP-1 in the medium for keratinized NHEK and in the conditioned medium of keratinized NHEK. The increase in CD54 and CD86 by changes in the medium type was higher than that by the NHEK secretion; therefore, it was found that the medium composition has a large effect on the evaluation index among the experimental parameters in the co-culture system. It is necessary to find the optimal medium for immunotoxicity assessment in the co-culture system.
Collapse
|
15
|
Phenotypic drug discovery: recent successes, lessons learned and new directions. Nat Rev Drug Discov 2022; 21:899-914. [DOI: 10.1038/s41573-022-00472-w] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/29/2022]
|
16
|
Brumm P, Fritschen A, Doß L, Dörsam E, Blaeser A. Fabrication of biomimetic networks using viscous fingering in flexographic printing. Biomed Mater 2022; 17. [PMID: 35579018 DOI: 10.1088/1748-605x/ac6b06] [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: 01/11/2022] [Accepted: 04/27/2022] [Indexed: 12/16/2022]
Abstract
Mammalian tissue comprises a plethora of hierarchically organized channel networks that serve as routes for the exchange of liquids, nutrients, bio-chemical cues or electrical signals, such as blood vessels, nerve fibers, or lymphatic conduits. Despite differences in function and size, the networks exhibit a similar, highly branched morphology with dendritic extensions. Mimicking such hierarchical networks represents a milestone in the biofabrication of tissues and organs. Work to date has focused primarily on the replication of the vasculature. Despite initial progress, reproducing such structures across scales and increasing biofabrication efficiency remain a challenge. In this work, we present a new biofabrication method that takes advantage of the viscous fingering phenomenon. Using flexographic printing, highly branched, inter-connective channel structures with stochastic, biomimetic distribution and dendritic extensions can be fabricated with unprecedented efficiency. Using gelatin (5%-35%) as resolvable sacrificial material, the feasability of the proposed method is demonstrated on the example of a vascular network. By selectively adjusting the printing velocity (0.2-1.5 m s-1), the anilox roller dip volume (4.5-24 ml m-2) as well as the shear viscosity of the printing material used (10-900 mPas), the width of the structures produced (30-400 µm) as well as their distance (200-600 µm) can be specifically determined. In addition to the flexible morphology, the high scalability (2500-25 000 mm2) and speed (1.5 m s-1) of the biofabrication process represents an important unique selling point. Printing parameters and hydrogel formulations are investigated and tuned towards a process window for controlled fabrication of channels that mimic the morphology of small blood vessels and capillaries. Subsequently, the resolvable structures were casted in a hydrogel matrix enabling bulk environments with integrated channels. The perfusability of the branched, inter-connective structures was successfully demonstrated. The fabricated networks hold great potential to enable nutrient supply in thick vascularized tissues or perfused organ-on-a-chip systems. In the future, the concept can be further optimized and expanded towards large-scale and cost-efficient biofabrication of vascular, lymphatic or neural networks for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Pauline Brumm
- Technical University of Darmstadt, Department of Mechanical Engineering, Institute of Printing Science and Technology, Magdalenenstr. 2, Darmstadt, 64289, Germany.,Collaborative Research Center (CRC) 1194-Interaction between Transport and Wetting Processes, Alarich-Weiss-Str. 10, Darmstadt, 64287, Germany
| | - Anna Fritschen
- Technical University of Darmstadt, Department of Mechanical Engineering, BioMedical Printing Technology, Magdalenenstr. 2, Darmstadt, 64289, Germany
| | - Lara Doß
- Technical University of Darmstadt, Department of Mechanical Engineering, BioMedical Printing Technology, Magdalenenstr. 2, Darmstadt, 64289, Germany
| | - Edgar Dörsam
- Technical University of Darmstadt, Department of Mechanical Engineering, Institute of Printing Science and Technology, Magdalenenstr. 2, Darmstadt, 64289, Germany.,Collaborative Research Center (CRC) 1194-Interaction between Transport and Wetting Processes, Alarich-Weiss-Str. 10, Darmstadt, 64287, Germany
| | - Andreas Blaeser
- Technical University of Darmstadt, Department of Mechanical Engineering, BioMedical Printing Technology, Magdalenenstr. 2, Darmstadt, 64289, Germany.,Technical University of Darmstadt, Centre for Synthetic Biology, Schnittspahnstr. 10, Darmstadt, 64287, Germany
| |
Collapse
|
17
|
Prince E, Kheiri S, Wang Y, Xu F, Cruickshank J, Topolskaia V, Tao H, Young EWK, McGuigan AP, Cescon DW, Kumacheva E. Microfluidic Arrays of Breast Tumor Spheroids for Drug Screening and Personalized Cancer Therapies. Adv Healthc Mater 2022; 11:e2101085. [PMID: 34636180 DOI: 10.1002/adhm.202101085] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/30/2021] [Indexed: 12/20/2022]
Abstract
One of the obstacles limiting progress in the development of effective cancer therapies is the shortage of preclinical models that capture the dynamic nature of tumor microenvironments. Interstitial flow strongly impacts tumor response to chemotherapy; however, conventional in vitro cancer models largely disregard this key feature. Here, a proof of principle microfluidic platform for the generation of large arrays of breast tumor spheroids that are grown under close-to-physiological flow in a biomimetic hydrogel is reported. This cancer spheroids-on-a-chip model is used for time- and labor-efficient studies of the effects of drug dose and supply rate on the chemosensitivity of breast tumor spheroids. The capability to grow large arrays of tumor spheroids from patient-derived cells of different breast cancer subtypes is shown, and the correlation between in vivo drug efficacy and on-chip spheroid drug response is demonstrated. The proposed platform can serve as an in vitro preclinical model for the development of personalized cancer therapies and effective screening of new anticancer drugs.
Collapse
Affiliation(s)
- Elisabeth Prince
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Sina Kheiri
- Department of Mechanical & Industrial Engineering University of Toronto 5 King's College Circle Toronto Ontario M5S 3G8 Canada
| | - Yihe Wang
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Fei Xu
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Jennifer Cruickshank
- Princess Margaret Cancer Centre University Health Network 610 University Ave Toronto Ontario M5G 2C1 Canada
| | - Valentina Topolskaia
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Huachen Tao
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
| | - Edmond W. K. Young
- Department of Mechanical & Industrial Engineering University of Toronto 5 King's College Circle Toronto Ontario M5S 3G8 Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto 164 College St Toronto Ontario M5S 3G9 Canada
| | - Alison. P. McGuigan
- Institute of Biomaterials and Biomedical Engineering University of Toronto 164 College St Toronto Ontario M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry University of Toronto 200 College St Toronto Ontario M5S 3E5 Canada
| | - David W. Cescon
- Princess Margaret Cancer Centre University Health Network 610 University Ave Toronto Ontario M5G 2C1 Canada
| | - Eugenia Kumacheva
- Department of Chemistry University of Toronto 80 St. George St Toronto Ontario M5P 2Y2 Canada
- Institute of Biomaterials and Biomedical Engineering University of Toronto 164 College St Toronto Ontario M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry University of Toronto 200 College St Toronto Ontario M5S 3E5 Canada
| |
Collapse
|
18
|
Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V, Vasconcelos MH. 3D Cell Culture Models as Recapitulators of the Tumor Microenvironment for the Screening of Anti-Cancer Drugs. Cancers (Basel) 2021; 14:190. [PMID: 35008353 PMCID: PMC8749977 DOI: 10.3390/cancers14010190] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Today, innovative three-dimensional (3D) cell culture models have been proposed as viable and biomimetic alternatives for initial drug screening, allowing the improvement of the efficiency of drug development. These models are gaining popularity, given their ability to reproduce key aspects of the tumor microenvironment, concerning the 3D tumor architecture as well as the interactions of tumor cells with the extracellular matrix and surrounding non-tumor cells. The development of accurate 3D models may become beneficial to decrease the use of laboratory animals in scientific research, in accordance with the European Union's regulation on the 3R rule (Replacement, Reduction, Refinement). This review focuses on the impact of 3D cell culture models on cancer research, discussing their advantages, limitations, and compatibility with high-throughput screenings and automated systems. An insight is also given on the adequacy of the available readouts for the interpretation of the data obtained from the 3D cell culture models. Importantly, we also emphasize the need for the incorporation of additional and complementary microenvironment elements on the design of 3D cell culture models, towards improved predictive value of drug efficacy.
Collapse
Affiliation(s)
- Mélanie A. G. Barbosa
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Biofabrication Group, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickevičiaus g 9, LT-44307 Kaunas, Lithuania;
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
19
|
Hu Q, Tang H, Yao Y, Liu S, Zhang H, Ramalingam M. Rapid fabrication of gelatin-based scaffolds with prevascularized channels for organ regeneration. Biomed Mater 2021; 16. [PMID: 33730706 DOI: 10.1088/1748-605x/abef7b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
One of the biggest hinders in tissue engineering over the last decades was the complexity of the prevascularized channels of the engineered scaffold, which was still lower than that of human tissues. Another relative trouble was lacking precision molding capability, which restricted the clinical applications of the huge engineered scaffold. In this study, a promising approach was proposed to prepare hydrogel scaffold with prevascularized channels by liquid bath printing, which chitosan/β-sodium glycerophosphate (CS/β-GP) severed as the ink hydrogel, and gelation/nanoscale bacterial cellulose (Gel/BC) acted as the supporting hydrogel. Here, the ink hydrogel was printed by a versatile nozzle and embedded in the supporting hydrogel. Ink hydrogel transformed into liquid effluent at low temperature after cross-linking of gelatin by microbial transglutaminase (mTG). No residual template was seen on the channel surface after template removal. This preparation had a high degree of freedom in the geometry of the channel, which was demonstrated by making various prevascularized channels including circular, branched, and tree-shaped networks. The molding accuracy of the channel was detected by studying the roundness of the cross-section of the molded hollow channel, and the effect of the mechanical properties by adding BC to supporting hydrogel was analyzed. Human umbilical vein endothelial cells (HUVECs) were injected into the aforementioned channels and formed confluent and homogeneous distribution on the surface of channels. Altogether, these results showed that this approach can construct hydrogel scaffold with complex and accurate molding prevascularized channels, and had great potential to resolve urgent vascularization issue of bulk tissue-engineering scaffold.
Collapse
Affiliation(s)
- Qingxi Hu
- Shanghai University, 99, , Shanghai, 200444, CHINA
| | - Haihu Tang
- Shanghai University, 99, , Shanghai, 200444, CHINA
| | - Yuan Yao
- Shanghai University, 99, , Shanghai, 200444, CHINA
| | - Suihong Liu
- Rapid Manufacturing Engineering Center, Shanghai University, No.99 Shangda Road, BaoShan District, Shanghai, China, Shanghai, 200444, CHINA
| | | | - Murugan Ramalingam
- Vellore Institute of Technology, Vandalur - Kelambakkam Road, Chennai , Vellore, Tamil Nadu, 632014, INDIA
| |
Collapse
|
20
|
Kadekar S, Barbe L, Stoddart M, Varghese OP, Tenje M, Mestres G. Effect of the Addition Frequency of 5-Azacytidine in Both Micro- and Macroscale Cultures. Cell Mol Bioeng 2021; 14:121-130. [PMID: 33633814 PMCID: PMC7878657 DOI: 10.1007/s12195-020-00654-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Human mesenchymal stem cells (hMSCs) have a great clinical potential for tissue regeneration purposes due to its multilineage capability. Previous studies have reported that a single addition of 5-azacytidine (5-AzaC) causes the differentiation of hMSCs towards a myocardial lineage. The aim of this work was to evaluate the effect of 5-AzaC addition frequency on hMSCs priming (i.e., indicating an early genetic differentiation) using two culture environments. METHODS hMSCs were supplemented with 5-AzaC while cultured in well plates and in microfluidic chips. The impact of 5-AzaC concentration (10 and 20 μM) and addition frequency (once, daily or continuously), as well as of culture period (2 or 5 days) on the genetic upregulation of PPARγ (adipocytes), PAX3 (myoblasts), SOX9 (chondrocytes) and RUNX2 (osteoblasts) was evaluated. RESULTS Daily delivering 5-AzaC caused a higher upregulation of PPARγ, SOX9 and RUNX2 in comparison to a single dose delivery, both under static well plates and dynamic microfluidic cultures. A particularly high gene expression of PPARγ (tenfold-change) could indicate priming of hMSCs towards adipocytes. CONCLUSIONS Both macro- and microscale cultures provided results with similar trends, where addition frequency of 5-AzaC was a crucial factor to upregulate several genes. Microfluidics technology was proven to be a suitable platform for the continuous delivery of a drug and could be used for screening purposes in tissue engineering research.
Collapse
Affiliation(s)
- Sandeep Kadekar
- Department of Chemistry-Ångström Laboratory, Uppsala University, 751 21 Uppsala, Sweden
| | - Laurent Barbe
- Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, Box 35, 751 03 Uppsala, Sweden
| | | | - Oommen P. Varghese
- Department of Chemistry-Ångström Laboratory, Uppsala University, 751 21 Uppsala, Sweden
| | - Maria Tenje
- Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, Box 35, 751 03 Uppsala, Sweden
| | - Gemma Mestres
- Department of Materials Science and Engineering, Science for Life Laboratory, Uppsala University, Box 35, 751 03 Uppsala, Sweden
| |
Collapse
|
21
|
García-Posadas L, Diebold Y. Three-Dimensional Human Cell Culture Models to Study the Pathophysiology of the Anterior Eye. Pharmaceutics 2020; 12:E1215. [PMID: 33333869 PMCID: PMC7765302 DOI: 10.3390/pharmaceutics12121215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023] Open
Abstract
In recent decades, the establishment of complex three-dimensional (3D) models of tissues has allowed researchers to perform high-quality studies and to not only advance knowledge of the physiology of these tissues but also mimic pathological conditions to test novel therapeutic strategies. The main advantage of 3D models is that they recapitulate the spatial architecture of tissues and thereby provide more physiologically relevant information. The eye is an extremely complex organ that comprises a large variety of highly heterogeneous tissues that are divided into two asymmetrical portions: the anterior and posterior segments. The anterior segment consists of the cornea, conjunctiva, iris, ciliary body, sclera, aqueous humor, and the lens. Different diseases in these tissues can have devastating effects. To study these pathologies and develop new treatments, the use of cell culture models is instrumental, and the better the model, the more relevant the results. Thus, the development of sophisticated 3D models of ocular tissues is a significant challenge with enormous potential. In this review, we present a comprehensive overview of the latest advances in the development of 3D in vitro models of the anterior segment of the eye, with a special focus on those that use human primary cells.
Collapse
Affiliation(s)
- Laura García-Posadas
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain;
| | - Yolanda Diebold
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, 47011 Valladolid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
22
|
Djeljadini S, Lohaus T, Gausmann M, Rauer S, Kather M, Krause B, Pich A, Möller M, Wessling M. Trypsin-Free Cultivation of 3D Mini-Tissues in an Adaptive Membrane Bioreactor. ACTA ACUST UNITED AC 2020; 4:e2000081. [PMID: 33089652 DOI: 10.1002/adbi.202000081] [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: 03/23/2020] [Revised: 08/11/2020] [Indexed: 11/07/2022]
Abstract
The production of large scaffold-free tissues is a key challenge in regenerative medicine. Nowadays, temperature-responsive polymers allow intact tissue harvesting without needing proteolytic enzymes. This method is limited to tissue culture plastic with limited upscaling capacity and plain process control. Here, a thermoresponsive hollow fiber membrane bioreactor is presented to produce large scaffold-free tissues. Intact tissues, rich in cell-to-cell connections and ECM, are harvested from a poly(N-vinylcaprolactam) microgel functionalized poly(ether sulfone)/poly(vinylpyrrolidone) hollow fiber membrane by a temperature shift. The harvested 3D tissues adhere in successive cultivation and exhibit high vitality for several days. The facile adsorptive coating waives the need for extensive surface treatment. The research is anticipated to be a starting point for upscaling the production of interconnected tissues enabling new opportunities in regenerative medicine, large-scale drug screening on physiological relevant tissues, and potentially opening new chances in cell-based therapies.
Collapse
Affiliation(s)
- Suzana Djeljadini
- Aachener Verfahrenstechnik, Chair of Chemical Process Engineering, RWTH Aachen University, Forckenbeckstrasse 51, Aachen, 52074, Germany
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, Aachen, 52074, Germany
| | - Theresa Lohaus
- Aachener Verfahrenstechnik, Chair of Chemical Process Engineering, RWTH Aachen University, Forckenbeckstrasse 51, Aachen, 52074, Germany
| | - Marcel Gausmann
- Aachener Verfahrenstechnik, Chair of Chemical Process Engineering, RWTH Aachen University, Forckenbeckstrasse 51, Aachen, 52074, Germany
| | - Sebastian Rauer
- Aachener Verfahrenstechnik, Chair of Chemical Process Engineering, RWTH Aachen University, Forckenbeckstrasse 51, Aachen, 52074, Germany
| | - Michael Kather
- Aachener Verfahrenstechnik, Chair of Chemical Process Engineering, RWTH Aachen University, Forckenbeckstrasse 51, Aachen, 52074, Germany
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, Aachen, 52074, Germany
| | - Bernd Krause
- Baxter International Inc., Research and Development, Holger-Crafoord-Straße 26, Hechingen, 72379, Germany
| | - Andrij Pich
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, Aachen, 52074, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, Aachen, 52074, Germany
| | - Martin Möller
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, Aachen, 52074, Germany
| | - Matthias Wessling
- Aachener Verfahrenstechnik, Chair of Chemical Process Engineering, RWTH Aachen University, Forckenbeckstrasse 51, Aachen, 52074, Germany
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, Aachen, 52074, Germany
| |
Collapse
|
23
|
Gundacker C, Ellinger I. The unique applicability of the human placenta to the Adverse Outcome Pathway (AOP) concept: the placenta provides fundamental insights into human organ functions at multiple levels of biological organization. Reprod Toxicol 2020; 96:273-281. [PMID: 32768559 DOI: 10.1016/j.reprotox.2020.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022]
Abstract
Despite the short lifespan of the human placenta, the proper formation and function of the organ is of crucial importance for fetal development. Placental dysfunction increases the risk of complications for mother and child during pregnancy and childbirth and beyond as it predisposes to fetal programming. The placenta is an upstream organ of the fetus. It performs the functions of fetal lungs, liver, intestines, kidneys and glands as long as these organs are not fully functional. Furthermore, it is the only human organ that is non-invasively available either after elective abortion or after birth. This is a crucial point given that the conceptual framework of Adverse Outcome Pathway (AOP) requires data on organ function. In vitro and ex vivo placental studies, combined with epidemiological and clinical data on pregnant women, newborns, and infants can uniquely cover all levels of information needed to develop new AOPs and complement existing AOPs related to reproductive toxicity and beyond. To stimulate further research in this area and to support researchers in future studies dealing with the development of AOPs related to the placenta, this review first gives a brief description of placental structure, placental development and relevant pregnancy diseases. The state of knowledge about the available placental models, their particularities and limitations are briefly discussed. Finally, the use of placental research for the development of AOPs is presented with an illustrative example.
Collapse
Affiliation(s)
- Claudia Gundacker
- Institute of Medical Genetics, Medical University Vienna, Vienna, Austria.
| | - Isabella Ellinger
- Institute for Pathophysiology and Allergy Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
24
|
Indirect co-culture of lung carcinoma cells with hyperthermia-treated mesenchymal stem cells influences tumor spheroid growth in a collagen-based 3-dimensional microfluidic model. Cytotherapy 2020; 23:25-36. [PMID: 32771259 DOI: 10.1016/j.jcyt.2020.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have paradoxically been reported to exert either pro- or anti-tumor effects in vitro. Hyperthermia, in combination with chemotherapy, has tumor-inhibiting effects; however, its role, together with MSCs, so far is not well understood. Furthermore, a lot of research is conducted using conventional 2-dimensional in vitro models that do not mimic the actual tumor microenvironment. AIM In light of this fact, an indirect method of co-culturing human amniotic membrane-derived MSCs (AMMSCs) with collagen-encapsulated human lung carcinoma cells (A549) was performed using a 3-dimensional (3D) tumor-on-chip device. METHODS The conditioned medium of AMMSCs (AMMSC-CM) or heat-treated AMMSCs (heat-AMMSC-CM) was utilized to create indirect co-culture conditions. Tumor spheroid growth characterization, immunocytochemistry and cytotoxicity assays, and anti-cancer peptide (P1) screening were performed to determine the effects of the conditioned medium. RESULTS The A549 cells cultured inside the 3D microfluidic chip developed into multicellular tumor spheroids over five days of culture. The AMMSC-CM, contrary to previous reports claiming its tumor-inhibiting potential, led to significant proliferation of tumor spheroids. Heat-AMMSC-CM led to reductions in both spheroid diameter and cell proliferation. The medium containing the P1 peptide was found to be the least cytotoxic to tumor spheroids in co-culture compared with the monoculture and heat-co-culture groups. CONCLUSIONS Hyperthermia, in combination with the anticancer peptide, exhibited highest cytotoxic effects. This study highlights the growing importance of 3D microfluidic tumor models for testing stem-cell-based and other anti-cancer therapies.
Collapse
|
25
|
Jessop ZM, Al-Sabah A, Simoes IN, Burnell SEA, Pieper IL, Thornton CA, Whitaker IS. Isolation and characterisation of nasoseptal cartilage stem/progenitor cells and their role in the chondrogenic niche. Stem Cell Res Ther 2020; 11:177. [PMID: 32408888 PMCID: PMC7222513 DOI: 10.1186/s13287-020-01663-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 02/21/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Since cartilage-derived stem/progenitor cells (CSPCs) were first identified in articular cartilage using differential adhesion to fibronectin, their self-renewal capacity and niche-specific lineage preference for chondrogenesis have propelled their application for cartilage tissue engineering. In many adult tissues, stem/progenitor cells are recognised to be involved in tissue homeostasis. However, the role of nasoseptal CSPCs has not yet been elucidated. Our aim was to isolate and characterise nasoseptal CSPCs alongside nasoseptal chondrocyte populations and determine chondrogenic capacity. METHODS Here, we isolated nasoseptal CSPCs using differential adhesion to fibronectin and assessed their colony forming efficiency, proliferation kinetics, karyotype and trilineage potential. CSPCs were characterised alongside non-fibronectin-adherent nasoseptal chondrocytes (DNCs) and cartilage-derived cells (CDCs, a heterogenous combination of DNCs and CSPCs) by assessing differences in gene expression profiles using PCR Stem Cell Array, immunophenotype using flow cytometry and chondrogencity using RT-PCR and histology. RESULTS CSPCs were clonogenic with increased gene expression of the neuroectodermal markers NCAM1 and N-Cadherin, as well as Cyclins D1 and D2, compared to DNCs. All three cell populations expressed recognised mesenchymal stem cell surface markers (CD29, CD44, CD73, CD90), yet only CSPCs and CDCs showed multilineage differentiation potential. CDC populations expressed significantly higher levels of type 2 collagen and bone morphogenetic protein 2 genes, with greater cartilage extracellular matrix secretion. When DNCs were cultured in isolation, there was reduced chondrogenicity and higher expression of type 1 collagen, stromal cell-derived factor 1 (SDF-1), CD73 and CD90, recognised markers of a fibroblast-like phenotype. CONCLUSIONS Fibronectin-adherent CSPCs demonstrate a unique gene expression profile compared to non-fibronectin-adherent DNCs. DNCs cultured in isolation, without CSPCs, express fibroblastic phenotype with reduced chondrogenicity. Mixed populations of stem/progenitor cells and chondrocytes were required for optimal chondrogenesis, suggesting that CSPCs may be required to retain phenotypic stability and chondrogenic potential of DNCs. Crosstalk between DNCs and CSPCs is proposed based on SDF-1 signalling.
Collapse
Affiliation(s)
- Zita M Jessop
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, SA2 8PP, UK.,Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK
| | - Ayesha Al-Sabah
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, SA2 8PP, UK
| | - Irina N Simoes
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, SA2 8PP, UK
| | - Stephanie E A Burnell
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, SA2 8PP, UK
| | - Ina Laura Pieper
- Calon Cardio-Technology Ltd, Institute of Life Sciences, Swansea, SA2 8PP, UK
| | - Catherine A Thornton
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, SA2 8PP, UK
| | - Iain S Whitaker
- Reconstructive Surgery and Regenerative Medicine Research Group, Institute of Life Sciences, Swansea University Medical School, Swansea, SA2 8PP, UK. .,Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, SA6 6NL, UK.
| |
Collapse
|
26
|
Thélu A, Catoire S, Kerdine-Römer S. Immune-competent in vitro co-culture models as an approach for skin sensitisation assessment. Toxicol In Vitro 2020; 62:104691. [DOI: 10.1016/j.tiv.2019.104691] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/05/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
|
27
|
Cutarelli A, Ghio S, Zasso J, Speccher A, Scarduelli G, Roccuzzo M, Crivellari M, Maria Pugno N, Casarosa S, Boscardin M, Conti L. Vertically-Aligned Functionalized Silicon Micropillars for 3D Culture of Human Pluripotent Stem Cell-Derived Cortical Progenitors. Cells 2019; 9:E88. [PMID: 31905823 PMCID: PMC7017050 DOI: 10.3390/cells9010088] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
Silicon is a promising material for tissue engineering since it allows to produce micropatterned scaffolding structures resembling biological tissues. Using specific fabrication methods, it is possible to build aligned 3D network-like structures. In the present study, we exploited vertically-aligned silicon micropillar arrays as culture systems for human iPSC-derived cortical progenitors. In particular, our aim was to mimic the radially-oriented cortical radial glia fibres that during embryonic development play key roles in controlling the expansion, radial migration and differentiation of cortical progenitors, which are, in turn, pivotal to the establishment of the correct multilayered cerebral cortex structure. Here we show that silicon vertical micropillar arrays efficiently promote expansion and stemness preservation of human cortical progenitors when compared to standard monolayer growth conditions. Furthermore, the vertically-oriented micropillars allow the radial migration distinctive of cortical progenitors in vivo. These results indicate that vertical silicon micropillar arrays can offer an optimal system for human cortical progenitors' growth and migration. Furthermore, similar structures present an attractive platform for cortical tissue engineering.
Collapse
Affiliation(s)
- Alessandro Cutarelli
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (J.Z.)
| | - Simone Ghio
- Fondazione Bruno Kessler-Center for Material and Microsystem, 38123 Trento, Italy; (S.G.); (M.C.)
| | - Jacopo Zasso
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (J.Z.)
| | - Alessandra Speccher
- Laboratory of Neural Development and Regeneration, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.S.); (S.C.)
| | - Giorgina Scarduelli
- Advanced Imaging Facility, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (G.S.); (M.R.)
| | - Michela Roccuzzo
- Advanced Imaging Facility, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (G.S.); (M.R.)
| | - Michele Crivellari
- Fondazione Bruno Kessler-Center for Material and Microsystem, 38123 Trento, Italy; (S.G.); (M.C.)
| | - Nicola Maria Pugno
- Laboratory of Bio-Inspired and Graphene Nanomechanics, Department of Civil, Environmental and Mechanical Engineering, University of Trento, 38123 Trento, Italy;
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
- Ket-Lab, Edoardo Amaldi Foundation, via del Politecnico snc, I-00133 Roma, Italy
| | - Simona Casarosa
- Laboratory of Neural Development and Regeneration, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.S.); (S.C.)
| | - Maurizio Boscardin
- Fondazione Bruno Kessler-Center for Material and Microsystem, 38123 Trento, Italy; (S.G.); (M.C.)
| | - Luciano Conti
- Laboratory of Stem Cell Biology, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, 38123 Trento, Italy; (A.C.); (J.Z.)
| |
Collapse
|
28
|
Booij TH, Price LS, Danen EHJ. 3D Cell-Based Assays for Drug Screens: Challenges in Imaging, Image Analysis, and High-Content Analysis. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2019; 24:615-627. [PMID: 30817892 PMCID: PMC6589915 DOI: 10.1177/2472555219830087] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 12/13/2022]
Abstract
The introduction of more relevant cell models in early preclinical drug discovery, combined with high-content imaging and automated analysis, is expected to increase the quality of compounds progressing to preclinical stages in the drug development pipeline. In this review we discuss the current switch to more relevant 3D cell culture models and associated challenges for high-throughput screening and high-content analysis. We propose that overcoming these challenges will enable front-loading the drug discovery pipeline with better biology, extracting the most from that biology, and, in general, improving translation between in vitro and in vivo models. This is expected to reduce the proportion of compounds that fail in vivo testing due to a lack of efficacy or to toxicity.
Collapse
Affiliation(s)
- Tijmen H. Booij
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- NEXUS Personalized Health Technologies, ETH Zürich, Switzerland
| | - Leo S. Price
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- OcellO B.V., Leiden, The Netherlands
| | - Erik H. J. Danen
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
29
|
Plasma-initiated polymerization of N-isopropylacrylamide and functionalized with dopamine for the adhesion to Hela cells. Polym Bull (Berl) 2019. [DOI: 10.1007/s00289-019-02784-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
30
|
Mestres G, Perez RA, D’Elía NL, Barbe L. Advantages of microfluidic systems for studying cell-biomaterial interactions—focus on bone regeneration applications. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab1033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
Mannhardt I, Warncke C, Trieu HK, Müller J, Eschenhagen T. Piezo-bending actuators for isometric or auxotonic contraction analysis of engineered heart tissue. J Tissue Eng Regen Med 2018; 13:3-11. [PMID: 30334614 DOI: 10.1002/term.2755] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/28/2018] [Accepted: 09/24/2018] [Indexed: 01/02/2023]
Abstract
Engineered heart tissue (EHT) has proven as valuable tool for disease modelling, drug safety screening, and cardiac repair. Especially in combination with the stem cell technology, these in vitro models of the human heart have generated interest not only of basic cardiovascular researchers but also of regulatory authorities responsible for drug safety. A main limitation of 3D-based assays for evaluating cardiotoxicity is their limited throughput. We integrated piezo-bending actuators in a 24-well system for the generation of strip-like rat and human EHT attached to hollow, elastic silicone posts. Muscle contractions of EHTs induced a measurable electrical current in the piezo-bending actuators that could be analysed for contraction amplitude, frequency, and contraction and relaxation kinetics. Compared with the standard video-optical analysis of contractile activity, the new system allows for (a) the analysis of several tissues in parallel, (b) switching between auxotonic and isometric contractions by inserting a stiff metal post in the silicone post opposing the piezo actuator, (c) continuous measurement over days with low data volume (megabyte), (d) automated measurement without the necessity of adjustment of tissue position for video-optical analysis, (e) reduced complexity and costs, (f) high sensitivity of contraction detection, (g) calculation of absolute contraction force, and (h) suitability for variable tissue geometries. The new set-up for contraction analysis based on piezo-bending actuators is a promising new method for the parallel screening of EHT for pharmacological drug effects and other applications of muscle tissue engineering (e.g., skeletal muscle engineering or cardiac repair).
Collapse
Affiliation(s)
- Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Christoph Warncke
- Institute of Microsystems Technology, Hamburg University of Technology, Hamburg, Germany
| | - Hoc Khiem Trieu
- Institute of Microsystems Technology, Hamburg University of Technology, Hamburg, Germany
| | - Jörg Müller
- Institute of Microsystems Technology, Hamburg University of Technology, Hamburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
32
|
Bale SS, Borenstein JT. Microfluidic Cell Culture Platforms to Capture Hepatic Physiology and Complex Cellular Interactions. Drug Metab Dispos 2018; 46:1638-1646. [PMID: 30115643 DOI: 10.1124/dmd.118.083055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/14/2018] [Indexed: 02/13/2025] Open
Abstract
Animal models such as rats and primates provide body-wide information for drug and metabolite responses, including organ-specific toxicity and any unforeseen side effects on other organs. Although effective in the drug-screening process, their translatability to humans is limited because of the lack of high concordance and correlation among enzymatic mechanisms, cellular mechanisms, and resulting toxicities. A significant mode of failure for safety prediction in drug screening is hepatotoxicity, resulting in ∼30% of all safety-related drug failures and withdrawals from the market. The liver is a multifunctional organ with diverse metabolic, secretory, and inflammatory response roles and is essential for maintaining key body functions. Conventional cell culture platforms (such as multiwell plate cultures) and metabolic enzyme systems (microsomes, cytochrome P450 enzymes) have been routinely used to assess drug pharmacokinetics and metabolism. However, current in vitro models often fail to recapitulate the complexity and dynamic nature of human tissues, imposing a heavy reliance on in vivo testing using preclinical species that have metabolic processes, disease mechanisms, and modes of toxicity distinct from humans. Recently, microphysiological systems (MPS) have gained attention as powerful tools with the potential to generate human-relevant information that can supplant and fill the gap of knowledge between preclinical animal models and simpler, conventional in vitro cell culture systems. Developments in microfabrication technologies for generating complex microfluidic systems, along with the ability to establish and maintain multicellular models to capture dynamic, human-relevant behavior, have provided new avenues to generate such physiologically relevant systems. These MPS platforms, when designed and developed with in vivo-derived design parameters, have the potential to capture key aspects and better mimic organ functionality. In this review, we discuss developments in microtechnologies for fabricating, establishing, and maintaining hepatic cell culture systems, with a specific focus on models that aim to capture in vivo physiology in vitro. By designing microscale systems to impart specific in vivo physiologic parameters, it is possible to create a dynamic system that can capture multiple aspects of the hepatic microenvironment, bringing us closer to a comprehensive in vitro testing platform for hepatic responses and toxicities.
Collapse
Affiliation(s)
- Shyam Sundhar Bale
- Cellular and Tissue Engineering, and Synthetic Biology and Bio-Instrumentation, Draper, Cambridge, Massachusetts
| | - Jeffrey T Borenstein
- Cellular and Tissue Engineering, and Synthetic Biology and Bio-Instrumentation, Draper, Cambridge, Massachusetts
| |
Collapse
|
33
|
Nie J, Gao Q, Wang Y, Zeng J, Zhao H, Sun Y, Shen J, Ramezani H, Fu Z, Liu Z, Xiang M, Fu J, Zhao P, Chen W, He Y. Vessel-on-a-chip with Hydrogel-based Microfluidics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802368. [PMID: 30307698 DOI: 10.1002/smll.201802368] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/18/2018] [Indexed: 05/20/2023]
Abstract
Hydrogel structures equipped with internal microchannels offer more in vivo-relevant models for construction of tissues and organs in vitro. However, currently used microfabrication methods of constructing microfluidic devices are not suitable for the handling of hydrogel. This study presents a novel method of fabricating hydrogel-based microfluidic chips by combining the casting and bonding processes. A twice cross-linking strategy is designed to obtain a bonding interface that has the same strength with the hydrogel bulk, which can be applied to arbitrary combinations of hydrogels. It is convenient to achieve the construction of hydrogel structures with channels in branched, spiral, serpentine, and multilayer forms. The experimental results show that the combination of gelatin and gelatin methacrylate (GelMA) owns the best biocompatibility and can promote cell functionalization. Based on these, a vessel-on-a-chip system with vascular function in both physiological and pathological situations is established, providing a promising model for further investigations such as vascularization, vascular inflammation, tissue engineering, and drug development. Taken together, a facile and cytocompatible approach is developed for engineering a user-defined hydrogel-based chip that can be potentially useful in developing vascularized tissue or organ models.
Collapse
Affiliation(s)
- Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yidong Wang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jiahui Zeng
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haiming Zhao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jian Shen
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hamed Ramezani
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhenliang Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhenjie Liu
- Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Peng Zhao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Wei Chen
- Children's Hospital Affiliated and Institute of Translational Medicine, Medical School, Zhejiang University, Hangzhou, 310029, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
34
|
In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2018; 198:228-249. [PMID: 30384974 PMCID: PMC7172914 DOI: 10.1016/j.biomaterials.2018.10.030] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Bacterial infections and antibiotic resistant bacteria have become a growing problem over the past decade. As a result, the Centers for Disease Control predict more deaths resulting from microorganisms than all cancers combined by 2050. Currently, many traditional models used to study bacterial infections fail to precisely replicate the in vivo bacterial environment. These models often fail to incorporate fluid flow, bio-mechanical cues, intercellular interactions, host-bacteria interactions, and even the simple inclusion of relevant physiological proteins in culture media. As a result of these inadequate models, there is often a poor correlation between in vitro and in vivo assays, limiting therapeutic potential. Thus, the urgency to establish in vitro and ex vivo systems to investigate the mechanisms underlying bacterial infections and to discover new-age therapeutics against bacterial infections is dire. In this review, we present an update of current in vitro and ex vivo models that are comprehensively changing the landscape of traditional microbiology assays. Further, we provide a comparative analysis of previous research on various established organ-disease models. Lastly, we provide insight on future techniques that may more accurately test new formulations to meet the growing demand of antibiotic resistant bacterial infections.
Collapse
|
35
|
Sensi F, D'Angelo E, D'Aronco S, Molinaro R, Agostini M. Preclinical three-dimensional colorectal cancer model: The next generation of in vitro drug efficacy evaluation. J Cell Physiol 2018; 234:181-191. [PMID: 30277557 DOI: 10.1002/jcp.26812] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
Colorectal cancer (CRC), the third most common cancer diagnosed in both men and women in the United States, shows a highly ineffective therapeutic management. In these years neither substantial improvements nor new therapeutic approaches have been provided to patients. Performing the early lead discovery phases of new cancer drugs in cellular models, resembling as far as possible the real in vivo tumor environment, may be more effective in predicting their future success in the later clinical phases. In this review, we critically describe the most representative bioengineered models for anticancer drug screening in CRC from the conventional two-dimensional models to the new-generation three-dimensional scaffold-based ones. The scaffold aims to replace the extracellular matrix, thus influencing the biomechanical, biochemical, and biological properties of cells and tissues. In this scenario, we believe that reconstitution of tumor condition is mandatory for an alternative in vitro methods to study cancer development and therapeutic strategies.
Collapse
Affiliation(s)
- Francesca Sensi
- Department of Women and Children Health, University of Padua, Padua, Italy.,Nano-Inspired Biomedicine Lab, Istituto di Ricerca Pediatrica - Città della Speranza, Padua, Italy
| | - Edoardo D'Angelo
- Nano-Inspired Biomedicine Lab, Istituto di Ricerca Pediatrica - Città della Speranza, Padua, Italy
| | - Sara D'Aronco
- Nano-Inspired Biomedicine Lab, Istituto di Ricerca Pediatrica - Città della Speranza, Padua, Italy.,Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Roberto Molinaro
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marco Agostini
- Nano-Inspired Biomedicine Lab, Istituto di Ricerca Pediatrica - Città della Speranza, Padua, Italy.,Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
36
|
Pupovac A, Senturk B, Griffoni C, Maniura-Weber K, Rottmar M, McArthur SL. Toward Immunocompetent 3D Skin Models. Adv Healthc Mater 2018. [PMID: 29542274 DOI: 10.1002/adhm.201701405] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
3D human skin models provide a platform for toxicity testing, biomaterials evaluation, and investigation of fundamental biological processes. However, the majority of current in vitro models lack an inflammatory system, vasculature, and other characteristics of native skin, indicating scope for more physiologically complex models. Looking at the immune system, there are a variety of cells that could be integrated to create novel skin models, but to do this effectively it is also necessary to understand the interface between skin biology and tissue engineering as well as the different roles the immune system plays in specific health and disease states. Here, a progress report on skin immunity and current immunocompetent skin models with a focus on construction methods is presented; scaffold and cell choice as well as the requirements of physiologically relevant models are elaborated. The wide range of technological and fundamental challenges that need to be addressed to successfully generate immunocompetent skin models and the steps currently being made globally by researchers as they develop new models are explored. Induced pluripotent stem cells, microfluidic platforms to control the model environment, and new real-time monitoring techniques capable of probing biochemical processes within the models are discussed.
Collapse
Affiliation(s)
- Aleta Pupovac
- Faculty of Science; Engineering and Technology; Swinburne University of Technology; Hawthorn Victoria 3122 Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO); Probing Biosystems Future Science Platform and Manufacturing; Clayton Victoria 3168 Australia
| | - Berna Senturk
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Chiara Griffoni
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Markus Rottmar
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Sally L. McArthur
- Faculty of Science; Engineering and Technology; Swinburne University of Technology; Hawthorn Victoria 3122 Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO); Probing Biosystems Future Science Platform and Manufacturing; Clayton Victoria 3168 Australia
| |
Collapse
|
37
|
Aljohani W, Ullah MW, Zhang X, Yang G. Bioprinting and its applications in tissue engineering and regenerative medicine. Int J Biol Macromol 2018; 107:261-275. [DOI: 10.1016/j.ijbiomac.2017.08.171] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 01/16/2023]
|
38
|
Carvalho MR, Reis RL, Oliveira JM. Mimicking the 3D biology of osteochondral tissue with microfluidic-based solutions: breakthroughs towards boosting drug testing and discovery. Drug Discov Today 2018; 23:711-718. [PMID: 29337200 DOI: 10.1016/j.drudis.2018.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/12/2017] [Accepted: 01/04/2018] [Indexed: 11/30/2022]
Abstract
The development of tissue-engineering (TE) solutions for osteochondral (OC) regeneration has been slowed by technical hurdles related to the recapitulation of their complex and hierarchical architecture. OC defects refer to damage of both the articular cartilage and the underlying subchondral bone. To repair an OC tissue defect, the complexity of the bone and cartilage must be considered. To help achieve this, microfluidics is converging with TE approaches to provide new treatment possibilities. Microfluidics uses precise micrometer-to-millimeter-scale fluid flows to achieve high-resolution and spatial and/or temporal control of the cell microenvironment, providing powerful tools for cell culturing. Herein, we overview the progress of microfluidics for developing 3D in vitro models of OC tissue, with a focus on cancer bone metastasis.
Collapse
Affiliation(s)
- Mariana R Carvalho
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga, 4805-017 Barco, Guimarães, Portugal
| | - Rui Luís Reis
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga, 4805-017 Barco, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Joaquim Miguel Oliveira
- 3Bs Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal; ICVS/3Bs - PT Government Associate Laboratory, Braga, 4805-017 Barco, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal.
| |
Collapse
|
39
|
A Method for Prostate and Breast Cancer Cell Spheroid Cultures Using Gelatin Methacryloyl-Based Hydrogels. Methods Mol Biol 2018; 1786:175-194. [PMID: 29786793 DOI: 10.1007/978-1-4939-7845-8_10] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Modern tissue engineering technologies have delivered tools to recreate a cell's naturally occurring niche in vitro and to investigate normal and pathological cell-cell and cell-niche interactions. Hydrogel biomaterials mimic crucial properties of native extracellular matrices, including mechanical support, cell adhesion sites and proteolytic degradability. As such, they are applied as 3D cell culture platforms to replicate tissue-like architectures observed in vivo, allowing physiologically relevant cell behaviors. Here we review bioengineered 3D approaches used for prostate and breast cancer. Furthermore, we describe the synthesis and use of gelatin methacryloyl-based hydrogels as in vitro 3D cancer model. This platform is used to engineer the microenvironments for prostate and breast cancer cells to study processes regulating spheroid formation, cell functions and responses to therapeutic compounds. Collectively, these bioengineered 3D approaches provide cell biologists with innovative pre-clinical tools that integrate the complexity of the disease seen in patients to advance our knowledge of cancer cell physiology and the contribution of a tumor's surrounding milieu.
Collapse
|
40
|
Ahadian S, Civitarese R, Bannerman D, Mohammadi MH, Lu R, Wang E, Davenport-Huyer L, Lai B, Zhang B, Zhao Y, Mandla S, Korolj A, Radisic M. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv Healthc Mater 2018; 7. [PMID: 29034591 DOI: 10.1002/adhm.201700506] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Significant advances in biomaterials, stem cell biology, and microscale technologies have enabled the fabrication of biologically relevant tissues and organs. Such tissues and organs, referred to as organ-on-a-chip (OOC) platforms, have emerged as a powerful tool in tissue analysis and disease modeling for biological and pharmacological applications. A variety of biomaterials are used in tissue fabrication providing multiple biological, structural, and mechanical cues in the regulation of cell behavior and tissue morphogenesis. Cells derived from humans enable the fabrication of personalized OOC platforms. Microscale technologies are specifically helpful in providing physiological microenvironments for tissues and organs. In this review, biomaterials, cells, and microscale technologies are described as essential components to construct OOC platforms. The latest developments in OOC platforms (e.g., liver, skeletal muscle, cardiac, cancer, lung, skin, bone, and brain) are then discussed as functional tools in simulating human physiology and metabolism. Future perspectives and major challenges in the development of OOC platforms toward accelerating clinical studies of drug discovery are finally highlighted.
Collapse
Affiliation(s)
- Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Robert Civitarese
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Rick Lu
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Erika Wang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Locke Davenport-Huyer
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Ben Lai
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Boyang Zhang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Serena Mandla
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| |
Collapse
|
41
|
Chen W, Liu X, Chen Q, Bao C, Zhao L, Zhu Z, Xu HHK. Angiogenic and osteogenic regeneration in rats via calcium phosphate scaffold and endothelial cell co-culture with human bone marrow mesenchymal stem cells (MSCs), human umbilical cord MSCs, human induced pluripotent stem cell-derived MSCs and human embryonic stem cell-derived MSCs. J Tissue Eng Regen Med 2017; 12:191-203. [PMID: 28098961 DOI: 10.1002/term.2395] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 10/03/2016] [Accepted: 01/09/2017] [Indexed: 02/05/2023]
Abstract
Angiogenesis is a limiting factor in regenerating large bone defects. The objective of this study was to investigate angiogenic and osteogenic effects of co-culture on calcium phosphate cement (CPC) scaffold using human umbilical vein endothelial cells (hUVECs) and mesenchymal stem cells (MSCs) from different origins for the first time. hUVECs were co-cultured with four types of cell: human umbilical cord MSCs (hUCMSCs), human bone marrow MSCs (hBMSCs) and MSCs from induced pluripotent stem cells (hiPSC-MSCs) and embryonic stem cells (hESC-MSCs). Constructs were implanted in 8 mm cranial defects of rats for 12 weeks. CPC without cells served as control 1. CPC with hBMSCs served as control 2. Microcapillary-like structures were successfully formed on CPC in vitro in all four co-cultured groups. Microcapillary lengths increased with time (p < 0.05). Osteogenic and angiogenic gene expressions were highly elevated and mineralization by co-cultured cells increased with time (p < 0.05). New bone amount and blood vessel density of co-cultured groups were much greater than controls (p < 0.05) in an animal study. hUVECs co-cultured with hUCMSCs, hiPSC-MSCs and hESC-MSCs achieved new bone and vessel density similar to hUVECs co-cultured with hBMSCs (p > 0.1). Therefore, hUCMSCs, hiPSC-MSCs and hESC-MSCs could serve as alternative cell sources to hBMSCs, which require an invasive procedure to harvest. In conclusion, this study showed for the first time that co-cultures of hUVECs with hUCMSCs, hiPSC-MSCs, hESC-MSCs and hBMSCs delivered via CPC scaffold achieved excellent osteogenic and angiogenic capabilities in vivo. The novel co-culture constructs are promising for bone reconstruction with improved angiogenesis for craniofacial/orthopaedic applications. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Wenchuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Xian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Qianmin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Liang Zhao
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA.,Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhimin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Periodontics and Prosthodontics, University of Maryland School of Dentistry, Baltimore, MD, USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Mechanical Engineering, University of Maryland at Baltimore County, Baltimore County, MD, USA
| |
Collapse
|
42
|
van den Broek LJ, Bergers LIJC, Reijnders CMA, Gibbs S. Progress and Future Prospectives in Skin-on-Chip Development with Emphasis on the use of Different Cell Types and Technical Challenges. Stem Cell Rev Rep 2017; 13:418-429. [PMID: 28536890 PMCID: PMC5486511 DOI: 10.1007/s12015-017-9737-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Understanding the healthy and diseased state of skin is important in many areas of basic and applied research. Although the field of skin tissue engineering has advanced greatly over the last years, current in vitro skin models still do not mimic the complexity of the human skin. Skin-on-chip and induced pluripotent stem cells (iPSC) might be key technologies to improve in vitro skin models. This review summarizes the state of the art of in vitro skin models with regard to cell sources (primary, cell line, iPSC) and microfluidic devices. It can be concluded that iPSC have the potential to be differentiated into many kinds of immunologically matched cells and skin-on-chip technology might lead to more physiologically relevant skin models due to the controlled environment, possible exchange of immune cells, and an increased barrier function. Therefore the combination of iPSC and skin-on-chip is expected to lead to superior healthy and diseased in vitro skin models.
Collapse
Affiliation(s)
| | | | | | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands.
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Pillet F, Gibot L, Madi M, Rols MP, Dague E. Importance of endogenous extracellular matrix in biomechanical properties of human skin model. Biofabrication 2017; 9:025017. [PMID: 28493850 DOI: 10.1088/1758-5090/aa6ed5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The physical and mechanical properties of cells modulate their behavior such proliferation rate, migration and extracellular matrix remodeling. In order to study cell behavior in a tissue-like environment in vitro, it is of utmost importance to develop biologically and physically relevant 3D cell models. Here, we characterized the physical properties of a single cell type growing in configurations of increasing complexity. From one human skin biopsy, primary dermal fibroblasts were isolated and seeded to give monolayer (2D model), spheroid (3D model poor in extracellular matrix) and tissue-engineered cell sheet (3D model rich in endogenous extracellular matrix). Living native human dermis tissue was used as a gold standard. Nanomechanical and viscoelastic properties at the cell scale were measured by atomic force microscopy (AFM) while biphoton microscopy allowed collagen detection by second harmonic generation and scanning electron microscopy helped in model morphological characterization. In all models, fibroblasts presented a similar typical elongated cell shape, with a cytoskeleton well-arranged along the long axis of the cell. However, elastic moduli of the tissue-engineered cell sheet and native dermis tissue were similar and statistically lower than monolayer and spheroid models. We successfully carried out AFM force measurements on 3D models such as spheroids and tissue-engineered cell sheets, as well as on living native human tissue. We demonstrated that a tissue-engineered dermal model recapitulates the mechanical properties of human native dermal tissue unlike the classically used monolayer and spheroid models. Furthermore, we give statistical evidence to indicate a correlation between cell mechanical properties and the presence of collagens in the models studied.
Collapse
Affiliation(s)
- Flavien Pillet
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | | | | | | |
Collapse
|
44
|
Oono M, Yamaguchi K, Rasyid A, Takano A, Tanaka M, Futai N. Reconfigurable microfluidic device with discretized sidewall. BIOMICROFLUIDICS 2017; 11:034103. [PMID: 28503247 PMCID: PMC5415406 DOI: 10.1063/1.4983148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/26/2017] [Indexed: 06/07/2023]
Abstract
Various microfluidic features, such as traps, have been used to manipulate flows, cells, and other particles within microfluidic systems. However, these features often become undesirable in subsequent steps requiring different fluidic configurations. To meet the changing needs of various microfluidic configurations, we developed a reconfigurable microfluidic channel with movable sidewalls using mechanically discretized sidewalls of laterally aligned rectangular pins. The user can deform the channel sidewall at any time after fabrication by sliding the pins. We confirmed that the flow resistance of the straight microchannel could be reversibly adjusted in the range of 101-105 Pa s/μl by manually displacing one of the pins comprising the microchannel sidewall. The reconfigurable microchannel also made it possible to manipulate flows and cells by creating a segmented patterned culture of COS-7 cells and a coculture of human umbilical vein endothelial cells (HUVECs) and human lung fibroblasts (hLFs) inside the microchannel. The reconfigurable microfluidic device successfully maintained a culture of COS-7 cells in a log phase throughout the entire period of 216 h. Furthermore, we performed a migration assay of cocultured HUVEC and hLF spheroids within one microchannel and observed their migration toward each other.
Collapse
Affiliation(s)
- Masahiro Oono
- Department of Mechanical Engineering, Graduate School of Engineering and Science, Shibaura Institute of Technology, 3-7-5 Toyosu, Koto-ku, Tokyo 135-8548, Japan
| | - Keisuke Yamaguchi
- Department of Mechanical Engineering, College of Engineering, Shibaura Institute of Technology, 3-7-5 Toyosu, Koto-ku, Tokyo 135-8548, Japan
| | - Amirul Rasyid
- Department of Mechanical Engineering, College of Engineering, Shibaura Institute of Technology, 3-7-5 Toyosu, Koto-ku, Tokyo 135-8548, Japan
| | - Atsushi Takano
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8 Somapah Rd, Singapore 487372
| | - Masato Tanaka
- Department of Materials and Life Sciences, School of Science and Engineering, Tokyo Denki University, Ishizaka, Hatoyama-machi, Hiki-gun, Saitama 350-0394, Japan
| | | |
Collapse
|
45
|
Ravichandran A, Liu Y, Teoh SH. Review: bioreactor design towards generation of relevant engineered tissues: focus on clinical translation. J Tissue Eng Regen Med 2017; 12:e7-e22. [PMID: 28374578 DOI: 10.1002/term.2270] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 07/13/2016] [Accepted: 07/19/2016] [Indexed: 12/27/2022]
Abstract
In tissue engineering and regenerative medicine, studies that utilize 3D scaffolds for generating voluminous tissues are mostly confined in the realm of in vitro research and preclinical animal model testing. Bioreactors offer an excellent platform to grow and develop 3D tissues by providing conditions that mimic their native microenvironment. Aligning the bioreactor development process with a focus on patient care will aid in the faster translation of the bioreactor technology to clinics. In this review, we discuss the various factors involved in the design of clinically relevant bioreactors in relation to their respective applications. We explore the functional relevance of tissue grafts generated by bioreactors that have been designed to provide physiologically relevant mechanical cues on the growing tissue. The review discusses the recent trends in non-invasive sensing of the bioreactor culture conditions. It provides an insight to the current technological advancements that enable in situ, non-invasive, qualitative and quantitative evaluation of the tissue grafts grown in a bioreactor system. We summarize the emerging trends in commercial bioreactor design followed by a short discussion on the aspects that hamper the 'push' of bioreactor systems into the commercial market as well as 'pull' factors for stakeholders to embrace and adopt widespread utility of bioreactors in the clinical setting. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- School of Chemical and Biomedical Engineering, 70 Nanyang Drive, Nanyang Technological University, Singapore, 637459, Singapore
| | - Yuchun Liu
- School of Chemical and Biomedical Engineering, 70 Nanyang Drive, Nanyang Technological University, Singapore, 637459, Singapore.,Academic Clinical Program (Research), National Dental Centre of Singapore, 5 Second Hospital Ave Singapore, 168938, Singapore
| | - Swee-Hin Teoh
- School of Chemical and Biomedical Engineering, 70 Nanyang Drive, Nanyang Technological University, Singapore, 637459, Singapore
| |
Collapse
|
46
|
Conzatti G, Cavalie S, Combes C, Torrisani J, Carrere N, Tourrette A. PNIPAM grafted surfaces through ATRP and RAFT polymerization: Chemistry and bioadhesion. Colloids Surf B Biointerfaces 2017; 151:143-155. [DOI: 10.1016/j.colsurfb.2016.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/25/2016] [Accepted: 12/07/2016] [Indexed: 12/23/2022]
|
47
|
Al-Himdani S, Jessop ZM, Al-Sabah A, Combellack E, Ibrahim A, Doak SH, Hart AM, Archer CW, Thornton CA, Whitaker IS. Tissue-Engineered Solutions in Plastic and Reconstructive Surgery: Principles and Practice. Front Surg 2017; 4:4. [PMID: 28280722 PMCID: PMC5322281 DOI: 10.3389/fsurg.2017.00004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/18/2017] [Indexed: 01/05/2023] Open
Abstract
Recent advances in microsurgery, imaging, and transplantation have led to significant refinements in autologous reconstructive options; however, the morbidity of donor sites remains. This would be eliminated by successful clinical translation of tissue-engineered solutions into surgical practice. Plastic surgeons are uniquely placed to be intrinsically involved in the research and development of laboratory engineered tissues and their subsequent use. In this article, we present an overview of the field of tissue engineering, with the practicing plastic surgeon in mind. The Medical Research Council states that regenerative medicine and tissue engineering “holds the promise of revolutionizing patient care in the twenty-first century.” The UK government highlighted regenerative medicine as one of the key eight great technologies in their industrial strategy worthy of significant investment. The long-term aim of successful biomanufacture to repair composite defects depends on interdisciplinary collaboration between cell biologists, material scientists, engineers, and associated medical specialties; however currently, there is a current lack of coordination in the field as a whole. Barriers to translation are deep rooted at the basic science level, manifested by a lack of consensus on the ideal cell source, scaffold, molecular cues, and environment and manufacturing strategy. There is also insufficient understanding of the long-term safety and durability of tissue-engineered constructs. This review aims to highlight that individualized approaches to the field are not adequate, and research collaboratives will be essential to bring together differing areas of expertise to expedite future clinical translation. The use of tissue engineering in reconstructive surgery would result in a paradigm shift but it is important to maintain realistic expectations. It is generally accepted that it takes 20–30 years from the start of basic science research to clinical utility, demonstrated by contemporary treatments such as bone marrow transplantation. Although great advances have been made in the tissue engineering field, we highlight the barriers that need to be overcome before we see the routine use of tissue-engineered solutions.
Collapse
Affiliation(s)
- Sarah Al-Himdani
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK
| | - Zita M Jessop
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK
| | - Ayesha Al-Sabah
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School , Swansea , UK
| | - Emman Combellack
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK
| | - Amel Ibrahim
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK; Institute of Child Health, University College London, London, UK
| | - Shareen H Doak
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; In Vitro Toxicology Group, Institute of Life Science, Swansea University Medical School, Swansea, UK
| | - Andrew M Hart
- Canniesburn Plastic Surgery Unit, Centre for Cell Engineering, University of Glasgow , Glasgow , UK
| | - Charles W Archer
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; Cartilage Biology Research Group, Institute of Life Science, Swansea University Medical School, Swansea, UK
| | - Catherine A Thornton
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; Human Immunology Group, Institute of Life Science, Swansea University Medical School, Swansea, UK
| | - Iain S Whitaker
- Reconstructive Surgery and Regenerative Medicine Research Group (ReconRegen), Institute of Life Science, Swansea University Medical School, Swansea, UK; The Welsh Centre for Burns and Plastic Surgery, Morriston Hospital, Swansea, UK
| |
Collapse
|
48
|
Iordache F, Constantinescu A, Andrei E, Amuzescu B, Halitzchi F, Savu L, Maniu H. Electrophysiology, immunophenotype, and gene expression characterization of senescent and cryopreserved human amniotic fluid stem cells. J Physiol Sci 2016; 66:463-476. [PMID: 27053101 PMCID: PMC10717473 DOI: 10.1007/s12576-016-0441-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/12/2016] [Indexed: 01/16/2023]
Abstract
We characterized human amniotic fluid stem cells (AFSC) in senescent cultures (6 weeks) versus cryopreserved cells using whole-cell patch-clamp, immunophenotyping, and differential gene expression profiling for senescence genes. We evidenced five ion current components (outward rectifier, A-type, inward rectifier, and big conductance Ca2+-dependent K+ currents, fast voltage-dependent Na+ currents). Senescent AFSC showed reduced expression of CD90, CD44, CD133, over 500-fold increase of interferon gamma and telomerase reverse transcriptase genes, increased cycle-dependent kinase 4 inhibitors, p53-binding protein 1, and decreased calreticulin and CD44. HLA-ABC immune expression was similar, and HLA-DR expression very low in both cell types. A subset of cryopreserved AFSC featured large inward rectifier K+ currents, voltage-dependent Na+ currents, and neural progenitor markers evidenced by immunophenotyping and RT-PCR. In all AFSC, in both culture conditions, at patch rupture the outward currents were very low, and they increased progressively over several minutes upon cytoplasm dialysis with pipette solution.
Collapse
Affiliation(s)
- Florin Iordache
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| | - Andrei Constantinescu
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| | - Eugen Andrei
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| | - Bogdan Amuzescu
- Department Biophysics and Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095, Bucharest, Romania.
| | - Ferdinand Halitzchi
- Department Biophysics and Physiology, Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095, Bucharest, Romania
| | - Lorand Savu
- Genetic Lab SRL, Cpt. Av. N. Drossu Str. 9, 012071, Bucharest, Romania
| | - Horia Maniu
- Flow Cytometry and Cell Therapy Laboratory, "N. Simionescu" Institute of Cellular Biology and Pathology, B.P. Hasdeu Str. 8, 050568, Bucharest, Romania
| |
Collapse
|
49
|
Microengineered platforms for co-cultured mesenchymal stem cells towards vascularized bone tissue engineering. Tissue Eng Regen Med 2016; 13:465-474. [PMID: 30603428 DOI: 10.1007/s13770-016-9080-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/08/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022] Open
Abstract
Bone defects are common disease requiring thorough treatments since the bone is a complex vascularized tissue that is composed of multiple cell types embedded within an intricate extracellular matrix (ECM). For past decades, tissue engineering using cells, proteins, and scaffolds has been suggested as one of the promising approaches for effective bone regeneration. Recently, many researchers have been interested in designing effective platform for tissue regeneration by orchestrating factors involved in microenvironment around tissues. Among factors affecting bone formation, vascularization during bone development and after minor insults via endochondral and intramembranous ossification is especially critical for the long-term support for functional bone. In order to create vascularized bone constructs, the interactions between human mesenchymal stem cells (MSCs) and endothelial cells (ECs) have been investigated using both direct and indirect co-culture studies. Recently, various culture methods including micropatterning techniques, three dimensional scaffolds, and microfluidics have been developed to create micro-engineered platforms that mimic the nature of vascularized bone formation, leading to the creation of functional bone structures. This review focuses on MSCs co-cultured with endothelial cells and microengineered platforms to determine the underlying interplay between co-cultured MSCs and vascularized bone constructs, which is ultimately necessary for adequate regeneration of bone defects.
Collapse
|
50
|
Rodil A, Laca A, Paredes B, Rendueles M, Meana Á, Díaz M. Gels prepared from egg yolk and its fractions for tissue engineering. Biotechnol Prog 2016; 32:1577-1583. [PMID: 27602804 DOI: 10.1002/btpr.2364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 06/30/2016] [Indexed: 11/09/2022]
Abstract
New biomaterials prepared from egg yolk and its main fractions (plasma and granules) have been developed for use in tissue engineering. Protein gels obtained via transglutaminase cross-linking were characterized by rheometry, texturometry and scanning electron microscopy. All the gels exhibited suitable physical and mechanical characteristics for use as potential biomaterials in skin regeneration. Specifically, results showed that these materials presented a compact, uniform structure, with granular gel being found to be the most resistant as well as the most elastic material. Accordingly, these gels were subsequently evaluated as scaffolds for murine fibroblast growth. The best results were obtained with granule gels. Not only adhesion and cell growth were detected when using these gels, but also continuous coatings of cells growing on their surface. These findings can be attributed to the higher protein content of this fraction and to the particular structure of its proteins. Thus, granules have proved to be an interesting potential raw material for scaffold development. © 2016 American Institute of Chemical Engineers Biotechnol. Prog., 32:1577-1583, 2016.
Collapse
Affiliation(s)
- Andrea Rodil
- Dept. of Chemical Engineering and Environmental Technology, University of Oviedo, C/Julián Clavería s/n, Oviedo, 33071, Spain
| | - Amanda Laca
- Dept. of Chemical Engineering and Environmental Technology, University of Oviedo, C/Julián Clavería s/n, Oviedo, 33071, Spain
| | - Benjamín Paredes
- Dept. of Chemical Engineering and Environmental Technology, University of Oviedo, C/Julián Clavería s/n, Oviedo, 33071, Spain
| | - Manuel Rendueles
- Dept. of Chemical Engineering and Environmental Technology, University of Oviedo, C/Julián Clavería s/n, Oviedo, 33071, Spain
| | - Álvaro Meana
- Community Center of Blood and Tissues of the Princedom of Asturias, C/Emilio Rodriguez Vigil s/n, Oviedo, 33006, Spain
| | - Mario Díaz
- Dept. of Chemical Engineering and Environmental Technology, University of Oviedo, C/Julián Clavería s/n, Oviedo, 33071, Spain
| |
Collapse
|