1001
|
Fabrication and Characterization of Scaffolds of Poly( ε-caprolactone)/Biosilicate® Biocomposites Prepared by Generative Manufacturing Process. Int J Biomater 2019; 2019:2131467. [PMID: 30853989 PMCID: PMC6377975 DOI: 10.1155/2019/2131467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/14/2019] [Indexed: 11/18/2022] Open
Abstract
Scaffolds of poly(ε-caprolactone) (PCL) and their biocomposites with 0, 1, 3, and 5 wt.% Biosilicate® were fabricated by the generative manufacturing process coupled with a vertical miniscrew extrusion head to application for restoration of bone tissue. Their morphological characterization indicated the designed 0°/90° architecture range of pore sizes and their interconnectivity is feasible for tissue engineering applications. Mechanical compression tests revealed an up to 57% increase in the stiffness of the scaffold structures with the addition of 1 to 5 wt.% Biosilicate® to the biocomposite. No toxicity was detected in the scaffolds tested by in vitro cell viability with MC3T3-E1 preosteoblast cell line. The results highlighted the potential application of scaffolds fabricated with poly(ε-caprolactone)/Biosilicate® to tissue engineering.
Collapse
|
1002
|
Schimke MM, Paul S, Tillmann K, Lepperdinger G, Stigler RG. Hard Tissue Augmentation of Aged Bone by Means of a Tin-Free PLLA-PCL Co-Polymer Exhibiting in vivo Anergy and Long-Term Structural Stability. Gerontology 2019; 65:174-185. [PMID: 30677770 DOI: 10.1159/000494798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Due to aging, tissue regeneration gradually declines. Contemporary strategies to promote tissue-specific regeneration, in particular in elderly patients, often include synthetic material apt for implantation primarily aiming at upholding body functions and regaining appropriate anatomical and functional integrity. OBJECTIVE Biomaterials suitable for complex reconstruction surgical procedures have to exert high physicochemical stability and biocompatibility. METHOD A polymer made of poly-L-lactic acid and poly-ε-caprolactone was synthesized by means of a novel tin-free catalytic process. The material was tested in a bioreactor-assisted perfusion culture and implanted in a sheep model for lateral augmentation of the mandible. Histological and volumetric evaluation was performed 3 and 6 months post-implantation. RESULTS After synthesis the material could be further refined by cryogrinding and sintering, thus yielding differently porous scaffolds that exhibited a firm and stable appearance. In perfusion culture, no disintegration was observed for extended periods of up to 7 weeks, while mesenchymal stromal cells readily attached to the material, steadily proliferated, and deposited extracellular calcium. The material was tested in vivo together with autologous bone marrow-derived stromal cells. Up to 6 months post-implantation, the material hardly changed in shape with composition also refraining from foreign body reactions. CONCLUSION Given the long-term shape stability in vivo, featuring imperceptible degradation and little scarring as well as exerting good compatibility to cells and surrounding tissues, this novel biomaterial is suitable as a space filler in large anatomical defects.
Collapse
|
1003
|
A three‐dimensional microfluidized liver system to assess hepatic drug metabolism and hepatotoxicity. Biotechnol Bioeng 2019; 116:1152-1163. [DOI: 10.1002/bit.26902] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022]
|
1004
|
Ray P, Alhalhooly L, Ghosh A, Choi Y, Banerjee S, Mallik S, Banerjee S, Quadir M. Size-Transformable, Multifunctional Nanoparticles from Hyperbranched Polymers for Environment-Specific Therapeutic Delivery. ACS Biomater Sci Eng 2019; 5:1354-1365. [DOI: 10.1021/acsbiomaterials.8b01608] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | | - Sushanta Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | |
Collapse
|
1005
|
Guo J, Zhao C, Yao R, Sui A, Sun L, Liu X, Wu S, Su Z, Li T, Liu S, Gao Y, Liu J, Feng X, Wang W, Zhao H, Cui Z, Li G, Meng F. 3D culture enhances chemoresistance of ALL Jurkat cell line by increasing DDR1 expression. Exp Ther Med 2019; 17:1593-1600. [PMID: 30783426 PMCID: PMC6364197 DOI: 10.3892/etm.2019.7153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/30/2018] [Indexed: 12/16/2022] Open
Abstract
Three dimensional (3D) culture has gradually become a research hotspot in the field of drug screening, stem cell research, and tissue engineering due to its more physiological-like morphology and function. In this study, we compared the differences of cell proliferation, population, protein expression and chemoresistance profiles between two dimensional (2D) and 3D culture of acute lymphoblastic leukemia (ALL) Jurkat cell line. Polycaprolactone (PCL) is used for 3D culture owing to its biochemical properties and compatibility. Culturing of ALL Jurkat cell line in collagen type I coated polycaprolactone scaffold for 168 h increased cell proliferation, attachment, as well as the drug resistance to cytarabine (Ara-C) and daunorubicin (DNR) without changing the original CD2+CD3+CD4+dimCD8−CD34−CD45+dim phenotype, compared to uncoated PCL scaffold and tissue culture plate systems. Molecularly, increased chemoresistance is associated with the upregulation of discoidin domain receptor 1 (DDR1) and transcription factor STAT3. Inhibition of DDR1 activity by DDR1-specific inhibitor DDR-IN-1 accelerated cell death in the presence of Ara-C, DNR or their combination. These results demonstrated that 3D culture enhances chemoresistance of ALL Jurkat cell line by increasing DDR1 expression. Importantly, the cell adhesion-mediated drug resistance induced by DDR1 in the scaffold was similar to the clinical situation, indicating the 3D culture of cancer cells recapitulate the in vivo tumor environment and this platform can be used as a promising pre-clinic drug-screen system.
Collapse
Affiliation(s)
- Jun Guo
- College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China.,Department of Hematology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Chunting Zhao
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Ruyong Yao
- Central Laboratory, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Aihua Sui
- Central Laboratory, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Lingjie Sun
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Xiaodan Liu
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Shaoling Wu
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Zhan Su
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Tianlan Li
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Shanshan Liu
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Yan Gao
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Jiaxiu Liu
- Central Laboratory, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Xianqi Feng
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Wei Wang
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Hongguo Zhao
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Zhongguang Cui
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Guanglun Li
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Fanjun Meng
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| |
Collapse
|
1006
|
Hui J, Pang S. Cell traction force in a confined microenvironment with double-sided micropost arrays. RSC Adv 2019; 9:8575-8584. [PMID: 35518671 PMCID: PMC9061871 DOI: 10.1039/c8ra10170a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/07/2019] [Indexed: 11/21/2022] Open
Abstract
Three-dimensional (3D) cell migrations are regulated by force interactions between cells and a 3D extracellular matrix (ECM). Mapping the 3D traction force generated by cells on the surrounding ECM with controlled confinement and contact area will be useful in understanding cell migration. In this study, double-sided micropost arrays were fabricated. The cell traction force was mapped by microposts on the top and bottom of opposing surfaces with a controlled separating distance to create different confinements. The density of micropost arrays was modified to investigate the effect of cell contact area on 3D traction force development. Using MC3T3-E1 osteoblastic cells, the leading traction force was found to increase with additional contact surface on the top. Summing force vectors on both surfaces, a large force imbalance was found from the leading to trailing regions for fast migrating cells. With 10 μm separation and densely arranged microposts, the traction force on the top surface was the largest at 28.6 ± 2.5 nN with the highest migration speed of 0.61 ± 0.07 μm min−1. Decreasing the density of the top micropost arrays resulted in a reduced traction force on the top and lower migration speed. With 15 μm separation, the cell traction force on the top and migration speed further decreased simultaneously. These results revealed traction force development on 3D ECM with varied degrees of confinement and contact area, which is important in regulating 3D cell migration. Double-sided micropost arrays to monitor three-dimensional cell traction force development over time on top and bottom surfaces with controlled confinement and contact area.![]()
Collapse
Affiliation(s)
- Jianan Hui
- Department of Electronic Engineering
- City University of Hong Kong
- China
- Center for Biosystems, Neuroscience, and Nanotechnology
- City University of Hong Kong
| | - Stella W. Pang
- Department of Electronic Engineering
- City University of Hong Kong
- China
- Center for Biosystems, Neuroscience, and Nanotechnology
- City University of Hong Kong
| |
Collapse
|
1007
|
Tellez-Gabriel M, Cochonneau D, Cadé M, Jubellin C, Heymann MF, Heymann D. Circulating Tumor Cell-Derived Pre-Clinical Models for Personalized Medicine. Cancers (Basel) 2018; 11:cancers11010019. [PMID: 30586936 PMCID: PMC6356998 DOI: 10.3390/cancers11010019] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022] Open
Abstract
The main cause of death from cancer is associated with the development of metastases, resulting from the inability of current therapies to cure patients at metastatic stages. Generating preclinical models to better characterize the evolution of the disease is thus of utmost importance, in order to implement effective new cancer biomarkers and therapies. Circulating Tumor Cells (CTCs) are good candidates for generating preclinical models, making it possible to follow up the spatial and temporal heterogeneity of tumor tissues. This method is a non-invasive liquid biopsy that can be obtained at any stage of the disease. It partially summarizes the molecular heterogeneity of the corresponding tumors at a given time. Here, we discuss the CTC-derived models that have been generated so far, from simplified 2D cultures to the most complex CTC-derived explants (CDX models). We highlight the challenges and strengths of these preclinical tools, as well as some of the recent studies published using these models.
Collapse
Affiliation(s)
- Marta Tellez-Gabriel
- RNA and Molecular Pathology Research Group, Department of Medical Biology, The Artic University of Norway, N-9037 Tromsø, Norway.
| | - Denis Cochonneau
- LabCT, Institut de Cancérologie de l'Ouest, CRCINA, Université d'Angers, 44805 Saint Herblain CEDEX, France.
| | - Marie Cadé
- INSERM, European Associated Laboratory "Sarcoma Research Unit", University of Nantes, 44035 Nantes, France.
| | - Camille Jubellin
- INSERM, European Associated Laboratory "Sarcoma Research Unit", University of Nantes, 44035 Nantes, France.
| | - Marie-Françoise Heymann
- LabCT, Institut de Cancérologie de l'Ouest, CRCINA, Université d'Angers, 44805 Saint Herblain CEDEX, France.
| | - Dominique Heymann
- LabCT, Institut de Cancérologie de l'Ouest, CRCINA, Université d'Angers, 44805 Saint Herblain CEDEX, France.
- INSERM, European Associated Laboratory "Sarcoma Research Unit", University of Nantes, 44035 Nantes, France.
- Department of Oncology & Metabolism, The Medical School, Beech Hill Road, Sheffield S10 2RX, UK.
| |
Collapse
|
1008
|
Santos SC, Custódio CA, Mano JF. Photopolymerizable Platelet Lysate Hydrogels for Customizable 3D Cell Culture Platforms. Adv Healthc Mater 2018; 7:e1800849. [PMID: 30387328 DOI: 10.1002/adhm.201800849] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/30/2018] [Indexed: 12/31/2022]
Abstract
3D cell culture platforms have emerged as a setting that resembles in vivo environments replacing the traditional 2D platforms. Over the recent years, an extensive effort has been made on the development of more physiologically relevant 3D cell culture platforms. Extracellular matrix-based materials have been reported as a bioactive and biocompatible support for cell culture. For example, human plasma derivatives have been extensively used in cell culture. Despite all the promising results, in most cases these types of materials have poor mechanical properties and poor stability in vitro. Here plasma-based hydrogels with increased stability are proposed. Platelet lysates are modified by addition of methacryloyl groups (PLMA) that polymerize in controlled geometries upon UV light exposure. The hydrogels could also generate porous scaffolds after lyophilization. The results show that PLMA materials have increased mechanical properties that can be easily adjusted by changing PLMA concentration or modification degree. Cells readily adhere, proliferate, and migrate, exhibiting high viability when encapsulated in PLMA hydrogels. The innovation potential of PLMA materials is based on the fact that it is a complete xeno-free solution for human cell culture, thus an effective alternative to the current gold standards for 3D cell culture based on animal products.
Collapse
Affiliation(s)
- Sara C. Santos
- Department of ChemistryCICECOUniversity of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - Catarina A. Custódio
- Department of ChemistryCICECOUniversity of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| | - João F. Mano
- Department of ChemistryCICECOUniversity of Aveiro Campus Universitário de Santiago 3810‐193 Aveiro Portugal
| |
Collapse
|
1009
|
Satpathy A, Datta P, Wu Y, Ayan B, Bayram E, Ozbolat IT. Developments with 3D bioprinting for novel drug discovery. Expert Opin Drug Discov 2018; 13:1115-1129. [PMID: 30384781 PMCID: PMC6494715 DOI: 10.1080/17460441.2018.1542427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023]
Abstract
Introduction: Although there have been significant contributions from the pharmaceutical industry to clinical practice, several diseases remain unconquered, with the discovery of new drugs remaining a paramount objective. The actual process of drug discovery involves many steps including pre-clinical and clinical testing, which are highly time- and resource-consuming, driving researchers to improve the process efficiency. The shift of modelling technology from two-dimensions (2D) to three-dimensions (3D) is one of such advancements. 3D Models allow for close mimicry of cellular interactions and tissue microenvironments thereby improving the accuracy of results. The advent of bioprinting for fabrication of tissues has shown potential to improve 3D culture models. Areas covered: The present review provides a comprehensive update on a wide range of bioprinted tissue models and appraise them for their potential use in drug discovery research. Expert opinion: Efficiency, reproducibility, and standardization are some impediments of the bioprinted models. Vascularization of the constructs has to be addressed in the near future. While much progress has already been made with several seminal works, the next milestone will be the commercialization of these models after due regulatory approval.
Collapse
Affiliation(s)
- Aishwarya Satpathy
- a Centre for Healthcare Science and Technology , Indian Institute of Engineering Science and Technology Shibpur , Howrah , India
| | - Pallab Datta
- a Centre for Healthcare Science and Technology , Indian Institute of Engineering Science and Technology Shibpur , Howrah , India
| | - Yang Wu
- b Engineering Science and Mechanics Department , Penn State University , University Park , PA , USA
- c The Huck Institutes of the Life Sciences, Penn State University , USA
| | - Bugra Ayan
- b Engineering Science and Mechanics Department , Penn State University , University Park , PA , USA
- c The Huck Institutes of the Life Sciences, Penn State University , USA
| | - Ertugrul Bayram
- d Medical Oncology Department , Agri State Hospital , Agri , Turkey
| | - Ibrahim T Ozbolat
- b Engineering Science and Mechanics Department , Penn State University , University Park , PA , USA
- c The Huck Institutes of the Life Sciences, Penn State University , USA
- e Biomedical Engineering Department , Penn State University , University Park , PA , USA
- f Materials Research Institute, Penn State University , USA
| |
Collapse
|
1010
|
Pihl AF, Offersgaard AF, Mathiesen CK, Prentoe J, Fahnøe U, Krarup H, Bukh J, Gottwein JM. High density Huh7.5 cell hollow fiber bioreactor culture for high-yield production of hepatitis C virus and studies of antivirals. Sci Rep 2018; 8:17505. [PMID: 30504788 PMCID: PMC6269495 DOI: 10.1038/s41598-018-35010-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/26/2018] [Indexed: 12/15/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection poses a serious global public health burden. Despite the recent development of effective treatments there is a large unmet need for a prophylactic vaccine. Further, antiviral resistance might compromise treatment efficiency in the future. HCV cell culture systems are typically based on Huh7 and derived hepatoma cell lines cultured in monolayers. However, efficient high cell density culture systems for high-yield HCV production and studies of antivirals are lacking. We established a system based on Huh7.5 cells cultured in a hollow fiber bioreactor in the presence or absence of bovine serum. Using an adapted chimeric genotype 5a virus, we achieved peak HCV infectivity and RNA titers of 7.6 log10 FFU/mL and 10.4 log10 IU/mL, respectively. Bioreactor derived HCV showed high genetic stability, as well as buoyant density, sensitivity to neutralizing antibodies AR3A and AR4A, and dependency on HCV co-receptors CD81 and SR-BI comparable to that of HCV produced in monolayer cell cultures. Using the bioreactor platform, treatment with the NS5A inhibitor daclatasvir resulted in HCV escape mediated by the NS5A resistance substitution Y93H. In conclusion, we established an efficient high cell density HCV culture system with implications for studies of antivirals and vaccine development.
Collapse
Affiliation(s)
- Anne F Pihl
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna F Offersgaard
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian K Mathiesen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jannick Prentoe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Krarup
- Section of Molecular Diagnostics, Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Judith M Gottwein
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
1011
|
Weiden J, Voerman D, Dölen Y, Das RK, van Duffelen A, Hammink R, Eggermont LJ, Rowan AE, Tel J, Figdor CG. Injectable Biomimetic Hydrogels as Tools for Efficient T Cell Expansion and Delivery. Front Immunol 2018; 9:2798. [PMID: 30546367 PMCID: PMC6279891 DOI: 10.3389/fimmu.2018.02798] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/13/2018] [Indexed: 12/22/2022] Open
Abstract
Biomaterial-based scaffolds are promising tools for controlled immunomodulation. They can be applied as three dimensional (3D) culture systems in vitro, whereas in vivo they may be used to dictate cellular localization and exert spatiotemporal control over cues presented to the immune system. As such, scaffolds can be exploited to enhance the efficacy of cancer immunotherapies such as adoptive T cell transfer, in which localization and persistence of tumor-specific T cells dictates treatment outcome. Biomimetic polyisocyanopeptide (PIC) hydrogels are polymeric scaffolds with beneficial characteristics as they display reversible thermally-induced gelation at temperatures above 16°C, which allows for their minimally invasive delivery via injection. Moreover, incorporation of azide-terminated monomers introduces functional handles that can be exploited to include immune cell-modulating cues. Here, we explore the potential of synthetic PIC hydrogels to promote the in vitro expansion and in vivo local delivery of pre-activated T cells. We found that PIC hydrogels support the survival and vigorous expansion of pre-stimulated T cells in vitro even at high cell densities, highlighting their potential as 3D culture systems for efficient expansion of T cells for their adoptive transfer. In particular, the reversible thermo-sensitive behavior of the PIC scaffolds favors straightforward recovery of cells. PIC hydrogels that were injected subcutaneously gelated instantly in vivo, after which a confined 3D structure was formed that remained localized for at least 4 weeks. Importantly, we noticed no signs of inflammation, indicating that PIC hydrogels are non-immunogenic. Cells co-delivered with PIC polymers were encapsulated within the scaffold in vivo. Cells egressed gradually from the PIC gel and migrated into distant organs. This confirms that PIC hydrogels can be used to locally deliver cells within a supportive environment. These results demonstrate that PIC hydrogels are highly promising for both the in vitro expansion and in vivo delivery of pre-activated T cells. Covalent attachment of biomolecules onto azide-functionalized PIC polymers provides the opportunity to steer the phenotype, survival or functional response of the adoptively transferred cells. As such, PIC hydrogels can be used as valuable tools to improve current adoptive T cell therapy strategies.
Collapse
Affiliation(s)
- Jorieke Weiden
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Dion Voerman
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Yusuf Dölen
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rajat K. Das
- Institute for Molecules and Materials, Radboud University, Nijmegen, Netherlands
- Materials Science Centre, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Anne van Duffelen
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Roel Hammink
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Loek J. Eggermont
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alan E. Rowan
- Institute for Molecules and Materials, Radboud University, Nijmegen, Netherlands
| | - Jurjen Tel
- Department of Biomedical Engineering, Laboratory of Immunoengineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Carl G. Figdor
- Department of Tumor Immunology, Oncode Institute, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
1012
|
Monteiro CF, Custódio CA, Mano JF. Three-Dimensional Osteosarcoma Models for Advancing Drug Discovery and Development. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Cátia F. Monteiro
- Department of Chemistry, CICECO; University of Aveiro, Campus Universitário de Santiago; 3810-193 Aveiro Portugal
| | - Catarina A. Custódio
- Department of Chemistry, CICECO; University of Aveiro, Campus Universitário de Santiago; 3810-193 Aveiro Portugal
| | - João F. Mano
- Department of Chemistry, CICECO; University of Aveiro, Campus Universitário de Santiago; 3810-193 Aveiro Portugal
| |
Collapse
|
1013
|
Randall MJ, Jüngel A, Rimann M, Wuertz-Kozak K. Advances in the Biofabrication of 3D Skin in vitro: Healthy and Pathological Models. Front Bioeng Biotechnol 2018; 6:154. [PMID: 30430109 PMCID: PMC6220074 DOI: 10.3389/fbioe.2018.00154] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/05/2018] [Indexed: 12/27/2022] Open
Abstract
The relevance for in vitro three-dimensional (3D) tissue culture of skin has been present for almost a century. From using skin biopsies in organ culture, to vascularized organotypic full-thickness reconstructed human skin equivalents, in vitro tissue regeneration of 3D skin has reached a golden era. However, the reconstruction of 3D skin still has room to grow and develop. The need for reproducible methodology, physiological structures and tissue architecture, and perfusable vasculature are only recently becoming a reality, though the addition of more complex structures such as glands and tactile corpuscles require advanced technologies. In this review, we will discuss the current methodology for biofabrication of 3D skin models and highlight the advantages and disadvantages of the existing systems as well as emphasize how new techniques can aid in the production of a truly physiologically relevant skin construct for preclinical innovation.
Collapse
Affiliation(s)
- Matthew J Randall
- Department of Health Science and Technology, Institute for Biomechanics, ETH Zürich, Zurich, Switzerland
| | - Astrid Jüngel
- Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, Switzerland
| | - Markus Rimann
- Competence Center TEDD, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Waedenswil, Switzerland.,Center for Cell Biology & Tissue Engineering, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Waedenswil, Switzerland
| | - Karin Wuertz-Kozak
- Department of Health Science and Technology, Institute for Biomechanics, ETH Zürich, Zurich, Switzerland.,Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (AU), Munich, Germany.,Department of Health Sciences, University of Potsdam, Potsdam, Germany
| |
Collapse
|
1014
|
Nunes AS, Costa EC, Barros AS, de Melo-Diogo D, Correia IJ. Establishment of 2D Cell Cultures Derived From 3D MCF-7 Spheroids Displaying a Doxorubicin Resistant Profile. Biotechnol J 2018; 14:e1800268. [PMID: 30242980 DOI: 10.1002/biot.201800268] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/14/2018] [Indexed: 01/09/2023]
Abstract
In vitro 3D cancer spheroids generally exhibit a drug resistance profile similar to that found in solid tumors. Due to this property, these models are an appealing for anticancer compounds screening. Nevertheless, the techniques and methods aimed for drug discovery are mostly standardized for cells cultured in 2D. The development of 2D cell culture models displaying a drug resistant profile is required to mimic the in vivo tumors, while the equipment, techniques, and methodologies established for conventional 2D cell cultures can continue to be employed in compound screening. In this work, the response of 3D-derived MCF-7 cells subsequently cultured in 2D in medium supplemented with glutathione (GSH) (antioxidant agent found in high levels in breast cancer tissues and a promoter of cancer cells resistance) to Doxorubicin (DOX) is evaluated. These cells demonstrated a resistance toward DOX closer to that displayed by 3D spheroids, which is higher than that exhibited by standard 2D cell cultures. In fact, the 50% inhibitory concentration (IC50 ) of DOX in 3D-derived MCF-7 cell cultures supplemented with GSH is about eight-times higher than that obtained for conventional 2D cell cultures (cultured without GSH), and is only about two-times lower than that attained for 3D MCF-7 spheroids (cultured without GSH). Further investigation revealed that this improved resistance of 3D-derived MCF-7 cells may result from their increased P-glycoprotein (P-gp) activity and reduced production of intracellular reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Ana S Nunes
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Elisabete C Costa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Andreia S Barros
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Duarte de Melo-Diogo
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Ilídio J Correia
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506, Covilhã, Portugal.,CIEPQF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, Polo II, 3030-790, Coimbra, Portugal
| |
Collapse
|
1015
|
Berg J, Hiller T, Kissner MS, Qazi TH, Duda GN, Hocke AC, Hippenstiel S, Elomaa L, Weinhart M, Fahrenson C, Kurreck J. Optimization of cell-laden bioinks for 3D bioprinting and efficient infection with influenza A virus. Sci Rep 2018; 8:13877. [PMID: 30224659 PMCID: PMC6141611 DOI: 10.1038/s41598-018-31880-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/28/2018] [Indexed: 01/12/2023] Open
Abstract
Bioprinting is a new technology, which arranges cells with high spatial resolution, but its potential to create models for viral infection studies has not yet been fully realized. The present study describes the optimization of a bioink composition for extrusion printing. The bioinks were biophysically characterized by rheological and electron micrographic measurements. Hydrogels consisting of alginate, gelatin and Matrigel were used to provide a scaffold for a 3D arrangement of human alveolar A549 cells. A blend containing 20% Matrigel provided the optimal conditions for spatial distribution and viability of the printed cells. Infection of the 3D model with a seasonal influenza A strain resulted in widespread distribution of the virus and a clustered infection pattern that is also observed in the natural lung but not in two-dimensional (2D) cell culture, which demonstrates the advantage of 3D printed constructs over conventional culture conditions. The bioink supported viral replication and proinflammatory interferon release of the infected cells. We consider our strategy to be paradigmatic for the generation of humanized 3D tissue models by bioprinting to study infections and develop new antiviral strategies.
Collapse
Affiliation(s)
- Johanna Berg
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355, Berlin, Germany
| | - Thomas Hiller
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355, Berlin, Germany
| | - Maya S Kissner
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355, Berlin, Germany
| | - Taimoor H Qazi
- Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Georg N Duda
- Berlin-Brandenburg Center for Regenerative Therapies & Berlin-Brandenburg School for Regenerative Therapies, Charité Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Andreas C Hocke
- Department of Internal Medicine/Infectious and Respiratory Diseases, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany, 10115, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine/Infectious and Respiratory Diseases, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany, 10115, Berlin, Germany
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Department of Organic Chemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Department of Organic Chemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Christoph Fahrenson
- Center for electron microscopy (ZELMI), Technische Universität Berlin, 10623, Berlin, Germany
| | - Jens Kurreck
- Institute of Biotechnology, Department of Applied Biochemistry, Technische Universität Berlin, 13355, Berlin, Germany.
| |
Collapse
|
1016
|
In Vitro Models for Studying Transport Across Epithelial Tissue Barriers. Ann Biomed Eng 2018; 47:1-21. [DOI: 10.1007/s10439-018-02124-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
|
1017
|
Rahimi C, Rahimi B, Padova D, Rooholghodos SA, Bienek DR, Luo X, Kaufman G, Raub CB. Oral mucosa-on-a-chip to assess layer-specific responses to bacteria and dental materials. BIOMICROFLUIDICS 2018; 12:054106. [PMID: 30310527 PMCID: PMC6158033 DOI: 10.1063/1.5048938] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/06/2018] [Indexed: 05/24/2023]
Abstract
The human oral mucosa hosts a diverse microbiome and is exposed to potentially toxic biomaterials from dental restoratives. Mucosal health is partly determined by cell and tissue responses to challenges such as dental materials and pathogenic bacteria. An in vitro model to rapidly determine potential layer-specific responses would lead to a better understanding of mucosal homeostasis and pathology. Therefore, this study aimed to develop a co-cultured microfluidic mucosal model on-a-chip to rapidly assess mucosal remodeling and the responses of epithelial and subepithelial layers to challenges typically found in the oral environment. A gingival fibroblast-laden collagen hydrogel was assembled in the central channel of a three-channel microfluidic chamber with interconnecting pores, followed by a keratinocyte layer attached to the collagen exposed in the pores. This configuration produced apical and subepithelial side channels capable of sustaining flow. Keratinocyte, fibroblast, and collagen densities were optimized to create a co-culture tissue-like construct stable over one week. Cells were stained and imaged with epifluorescence microscopy to confirm layer characteristics. As proof-of-concept, the mucosal construct was exposed separately to a dental monomer, 2-hydroxylethyl methacrylate (HEMA), and the oral bacteria Streptococcus mutans. Exposure to HEMA lowered mucosal cell viability, while exposure to the bacteria lowered trans-epithelial electrical resistance. These findings suggest that the oral mucosa-on-a-chip is useful for studying oral mucosal interactions with bacteria and biomaterials with a histology-like view of the tissue layers.
Collapse
Affiliation(s)
- Christopher Rahimi
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, USA
| | - Benjamin Rahimi
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, USA
| | - Dominic Padova
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, USA
| | - Seyed A. Rooholghodos
- Department of Mechanical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, USA
| | - Diane R. Bienek
- ADA Foundation, Volpe Research Center, 100 Bureau Drive, Stop #8546, Gaithersburg, Maryland 20899, USA
| | - Xiaolong Luo
- Department of Mechanical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, USA
| | - Gili Kaufman
- ADA Foundation, Volpe Research Center, 100 Bureau Drive, Stop #8546, Gaithersburg, Maryland 20899, USA
| | - Christopher B. Raub
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, District of Columbia 20064, USA
| |
Collapse
|
1018
|
Dixit C, Kadimisetty K, Rusling J. 3D-printed miniaturized fluidic tools in chemistry and biology. Trends Analyt Chem 2018; 106:37-52. [PMID: 32296252 PMCID: PMC7158885 DOI: 10.1016/j.trac.2018.06.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
3D printing (3DP), an additive manufacturing (AM) approach allowing for rapid prototyping and decentralized fabrication on-demand, has become a common method for creating parts or whole devices. The wide scope of the AM extends from organized sectors of construction, ornament, medical, and R&D industries to individual explorers attributed to the low cost, high quality printers along with revolutionary tools and polymers. While progress is being made but big manufacturing challenges are still there. Considering the quickly shifting narrative towards miniaturized analytical systems (MAS) we focus on the development/rapid prototyping and manufacturing of MAS with 3DP, and application dependent challenges in engineering designs and choice of the polymeric materials and provide an exhaustive background to the applications of 3DP in biology and chemistry. This will allow readers to perceive the most important features of AM in creating (i) various individual and modular components, and (ii) complete integrated tools.
Collapse
Affiliation(s)
- C.K. Dixit
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, United States
| | - K. Kadimisetty
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, United States
| | - J. Rusling
- Department of Chemistry, University of Connecticut, Storrs, CT 06269-3060, United States
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269-3136, United States
- Department of Surgery and Neag Cancer Centre, UConn Health, Farmington, CT 06030, United States
- School of Chemistry, National University of Ireland at Galway, Galway, Ireland
| |
Collapse
|
1019
|
Zhu S, Ehnert S, Rouß M, Häussling V, Aspera-Werz RH, Chen T, Nussler AK. From the Clinical Problem to the Basic Research-Co-Culture Models of Osteoblasts and Osteoclasts. Int J Mol Sci 2018; 19:2284. [PMID: 30081523 PMCID: PMC6121694 DOI: 10.3390/ijms19082284] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Bone tissue undergoes constant remodeling and healing when fracture happens, in order to ensure its structural integrity. In order to better understand open biological and clinical questions linked to various bone diseases, bone cell co-culture technology is believed to shed some light into the dark. Osteoblasts/osteocytes and osteoclasts dominate the metabolism of bone by a multitude of connections. Therefore, it is widely accepted that a constant improvement of co-culture models with both cell types cultured on a 3D scaffold, is aimed to mimic an in vivo environment as closely as possible. Although in recent years a considerable knowledge of bone co-culture models has been accumulated, there are still many open questions. We here try to summarize the actual knowledge and address open questions.
Collapse
Affiliation(s)
- Sheng Zhu
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Marc Rouß
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Victor Häussling
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Romina H Aspera-Werz
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Tao Chen
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Andreas K Nussler
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| |
Collapse
|
1020
|
An S. The emerging role of extracellular Ca
2+
in osteo/odontogenic differentiation and the involvement of intracellular Ca
2+
signaling: From osteoblastic cells to dental pulp cells and odontoblasts. J Cell Physiol 2018; 234:2169-2193. [DOI: 10.1002/jcp.27068] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Shaofeng An
- Department of Operative Dentistry and EndodonticsGuanghua School of Stomatology, Hospital of Stomatology, Sun Yat‐sen UniversityGuangzhou China
- Guangdong Province Key Laboratory of StomatologySun Yat‐Sen UniversityGuangzhou China
| |
Collapse
|
1021
|
Cui H, Miao S, Esworthy T, Zhou X, Lee SJ, Liu C, Yu ZX, Fisher JP, Mohiuddin M, Zhang LG. 3D bioprinting for cardiovascular regeneration and pharmacology. Adv Drug Deliv Rev 2018; 132:252-269. [PMID: 30053441 PMCID: PMC6226324 DOI: 10.1016/j.addr.2018.07.014] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/22/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Compared to traditional therapeutic strategies, three-dimensional (3D) bioprinting is one of the most advanced techniques for creating complicated cardiovascular implants with biomimetic features, which are capable of recapitulating both the native physiochemical and biomechanical characteristics of the cardiovascular system. The present review provides an overview of the cardiovascular system, as well as describes the principles of, and recent advances in, 3D bioprinting cardiovascular tissues and models. Moreover, this review will focus on the applications of 3D bioprinting technology in cardiovascular repair/regeneration and pharmacological modeling, further discussing current challenges and perspectives.
Collapse
Affiliation(s)
- Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Shida Miao
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Chengyu Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742, USA
| | | | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
1022
|
Pajorova J, Bacakova M, Musilkova J, Broz A, Hadraba D, Lopot F, Bacakova L. Morphology of a fibrin nanocoating influences dermal fibroblast behavior. Int J Nanomedicine 2018; 13:3367-3380. [PMID: 29922057 PMCID: PMC5997127 DOI: 10.2147/ijn.s162644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Our study focuses on the fabrication of appropriate scaffolds for skin wound healing. This research brings valuable insights into the molecular mechanisms of adhesion, proliferation, and control of cell behavior through the extracellular matrix represented by synthetic biodegradable nanofibrous membranes coated by biomolecules. METHODS Nanofibrous polylactic acid (PLA) membranes were prepared by a needle-less electrospinning technology. These membranes were coated with fibrin according to two preparation protocols, and additionally they were coated with fibronectin in order to increase the cell affinity for colonizing the PLA membranes. The adhesion, growth, and extracellular matrix protein production of neonatal human dermal fibroblasts were evaluated on the nanofibrous membranes. RESULTS Our results showed that fibrin-coated membranes improved the adhesion and proliferation of human dermal fibroblasts. The morphology of the fibrin nanocoating seems to be crucial for the adhesion of fibroblasts, and consequently for their phenotypic maturation. Fibrin either covered the individual fibers in the membrane (F1 nanocoating), or covered the individual fibers and also formed a fine homogeneous nanofibrous mesh on the surface of the membrane (F2 nanocoating), depending on the mode of fibrin preparation. The fibroblasts on the membranes with the F1 nanocoating remained in their typical spindle-like shape. However, the cells on the F2 nanocoating were spread mostly in a polygon-like shape, and their proliferation was significantly higher. Fibronectin formed an additional mesh attached to the surface of the fibrin mesh, and further enhanced the cell adhesion and growth. The relative gene expression and protein production of collagen I and fibronectin were higher on the F2 nanocoating than on the F1 nanocoating. CONCLUSION A PLA membrane coated with a homogeneous fibrin mesh seems to be promising for the construction of temporary full-thickness skin tissue substitutes.
Collapse
Affiliation(s)
- Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Antonin Broz
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Daniel Hadraba
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Anatomy and Biomechanics, Faculty of Physical Education and Sport, Charles University, Prague, Czech Republic
| | - Frantisek Lopot
- Department of Anatomy and Biomechanics, Faculty of Physical Education and Sport, Charles University, Prague, Czech Republic
| | - Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
1023
|
Nano-scale microfluidics to study 3D chemotaxis at the single cell level. PLoS One 2018; 13:e0198330. [PMID: 29879160 PMCID: PMC5991685 DOI: 10.1371/journal.pone.0198330] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/17/2018] [Indexed: 11/19/2022] Open
Abstract
Directed migration of cells relies on their ability to sense directional guidance cues and to interact with pericellular structures in order to transduce contractile cytoskeletal- into mechanical forces. These biomechanical processes depend highly on microenvironmental factors such as exposure to 2D surfaces or 3D matrices. In vivo, the majority of cells are exposed to 3D environments. Data on 3D cell migration are mostly derived from intravital microscopy or collagen-based in vitro assays. Both approaches offer only limited controllability of experimental conditions. Here, we developed an automated microfluidic system that allows positioning of cells in 3D microenvironments containing highly controlled diffusion-based chemokine gradients. Tracking migration in such gradients was feasible in real time at the single cell level. Moreover, the setup allowed on-chip immunocytochemistry and thus linking of functional with phenotypical properties in individual cells. Spatially defined retrieval of cells from the device allows down-stream off-chip analysis. Using dendritic cells as a model, our setup specifically allowed us for the first time to quantitate key migration characteristics of cells exposed to identical gradients of the chemokine CCL19 yet placed on 2D vs in 3D environments. Migration properties between 2D and 3D migration were distinct. Morphological features of cells migrating in an in vitro 3D environment were similar to those of cells migrating in animal tissues, but different from cells migrating on a surface. Our system thus offers a highly controllable in vitro-mimic of a 3D environment that cells traffic in vivo.
Collapse
|
1024
|
Dixon EE, Woodward OM. Three-dimensional in vitro models answer the right questions in ADPKD cystogenesis. Am J Physiol Renal Physiol 2018; 315:F332-F335. [PMID: 29693448 DOI: 10.1152/ajprenal.00126.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Novel technologies, new understanding of the basement membrane composition, and better comprehension of the embryonic development of the mammalian kidney have led to explosive growth in the use of three-dimensional in vitro models to study a range of human disease pathologies (Clevers H. Cell 165: 1586-1597, 2016; Shamir ER, Ewald AJ. Nat Rev Mol Cell Biol 15: 647-664, 2014). The development of these effective model systems represents a new tool to study the progressive cystogenesis of autosomal dominant polycystic kidney disease (ADPKD). ADPKD is a prevalent and complex monogenetic disease, characterized by the pathological formation of fluid fill cysts in renal tissue (Grantham JJ, Mulamalla S, Swenson-Fields KI. Nat Rev Nephrol 7: 556-566, 2011; Takiar V, Caplan MJ. Biochim Biophys Acta 1812: 1337-1343, 2011). ADPKD cystogenesis is attributed to loss of function mutations in either PKD1 or PKD2, which encode for two transmembrane proteins, polycystin-1 and polycystin-2, and progresses with loss of both copies of either gene through a proposed two-hit mechanism with secondary somatic mutations (Delmas P, Padilla F, Osorio N, Coste B, Raoux M, Crest M. Biochem Biophys Res Commun 322: 1374-1383, 2004; Pei Y, Watnick T, He N, Wang K, Liang Y, Parfrey P, Germino G, St George-Hyslop P. Am Soc Nephrol 10: 1524-1529, 1999; Wu G, D'Agati V, Cai Y, Markowitz G, Park JH, Reynolds DM, Maeda Y, Le TC, Hou H Jr, Kucherlapati R, Edelmann W, Somlo S. Cell 93: 177-188, 1998). The exaggerated consequences of large fluid filled cysts result in fibrosis and nephron injury, leading initially to functional compensation but ultimately to dysfunction (Grantham JJ. Am J Kidney Dis 28: 788-803, 1996; Norman J. Biochim Biophys Acta 1812: 1327-1336, 2011; Song CJ, Zimmerman KA, Henke SJ, Yoder BK. Results Probl Cell Differ 60: 323-344, 2017). The complicated disease progression has scattered focus and resources across the spectrum of ADPKD research.
Collapse
Affiliation(s)
- Eryn E Dixon
- Department of Physiology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
1025
|
Daskalaki E, Pillon NJ, Krook A, Wheelock CE, Checa A. The influence of culture media upon observed cell secretome metabolite profiles: The balance between cell viability and data interpretability. Anal Chim Acta 2018; 1037:338-350. [PMID: 30292310 DOI: 10.1016/j.aca.2018.04.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022]
Abstract
The application of metabolomics to investigating the cell secretome has garnered popularity owing to the method's large-scale data output, biochemical insight, and prospects for novel target compound discovery. However, there are no standardized protocols for the use of cell growth media, a factor that can exert profound effects upon the detected metabolites, and thus in the interpretability of the resulting data. Herein, we applied a liquid chromatography-high resolution mass spectrometry-based metabolomics approach to examine the influence of 5 different media combinations upon the obtained secretome of two phenotypically different cell lines: human embryonic kidney cells (HEK293) and L6 rat muscle cells. These media combinations were, M1: Medium 199, M2: Medium 199 + 2% fetal bovine serum (FBS), M3: Dulbecco's Modified Eagle's Medium (DMEM), M4: DMEM + 2% FBS and M5: Krebs-Henseleit Modified Buffer (KHB). The effect of incubation (37 °C) vs. refrigeration (4 °C) on DMEM medium over a 24 h period was also investigated. Results were validated for a selected panel of 5 metabolites measured from an additional cell culture experiment. Metabolomics identified a total of 53 polar metabolites that exhibited differential patterns on a cell type- and medium-specific basis. We observed that choice of media was the primary contributor to the secreted metabolite profile detected. The addition of FBS resulted in unique detected metabolites, compared to media-only controls (M199 and DMEM alone). Glutamine and pyroglutamate were more abundant in incubated relative to refrigerated DMEM medium. The overall metabolic pattern of the metabolites from the targeted approach matched with that exhibited across M1-M5 of the metabolomics experiment, and aided in further identifying the presence of compounds that were below the limit of detection in metabolomics. Based upon these findings, we highlight the following considerations in designing a cell secretome-based metabolite profiling experiment: (1) multiple media combinations (with and without FBS) should be tested for each cell line to be investigated; (2) cell-free media combinations should be plated separately, and incubated/treated in the same experimental conditions as the cells; and (3) a compromise between cell death and metabolite detection should be identified in order to avoid batch-specific contributions from FBS supplementation.
Collapse
Affiliation(s)
- Evangelia Daskalaki
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, SE-171 77 Stockholm, Sweden
| | - Nicolas J Pillon
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, von Eulers Väg 4a, SE-171 77 Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Integrative Physiology, Karolinska Institutet, von Eulers Väg 4a, SE-171 77 Stockholm, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, SE-171 77 Stockholm, Sweden.
| | - Antonio Checa
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
1026
|
Progression of conventional hepatic cell culture models to bioengineered HepG2 cells for evaluation of herbal bioactivities. Biotechnol Lett 2018; 40:881-893. [DOI: 10.1007/s10529-018-2547-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/30/2018] [Indexed: 12/26/2022]
|
1027
|
Disease Modeling Using 3D Organoids Derived from Human Induced Pluripotent Stem Cells. Int J Mol Sci 2018; 19:ijms19040936. [PMID: 29561796 PMCID: PMC5979503 DOI: 10.3390/ijms19040936] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 12/12/2022] Open
Abstract
The rising interest in human induced pluripotent stem cell (hiPSC)-derived organoid culture has stemmed from the manipulation of various combinations of directed multi-lineage differentiation and morphogenetic processes that mimic organogenesis. Organoids are three-dimensional (3D) structures that are comprised of multiple cell types, self-organized to recapitulate embryonic and tissue development in vitro. This model has been shown to be superior to conventional two-dimensional (2D) cell culture methods in mirroring functionality, architecture, and geometric features of tissues seen in vivo. This review serves to highlight recent advances in the 3D organoid technology for use in modeling complex hereditary diseases, cancer, host–microbe interactions, and possible use in translational and personalized medicine where organoid cultures were used to uncover diagnostic biomarkers for early disease detection via high throughput pharmaceutical screening. In addition, this review also aims to discuss the advantages and shortcomings of utilizing organoids in disease modeling. In summary, studying human diseases using hiPSC-derived organoids may better illustrate the processes involved due to similarities in the architecture and microenvironment present in an organoid, which also allows drug responses to be properly recapitulated in vitro.
Collapse
|
1028
|
Qiao H, Tang T. Engineering 3D approaches to model the dynamic microenvironments of cancer bone metastasis. Bone Res 2018; 6:3. [PMID: 29507817 PMCID: PMC5826951 DOI: 10.1038/s41413-018-0008-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 12/01/2017] [Accepted: 12/27/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer metastasis to bone is a three-dimensional (3D), multistep, dynamic process that requires the sequential involvement of three microenvironments, namely, the primary tumour microenvironment, the circulation microenvironment and the bone microenvironment. Engineered 3D approaches allow for a vivid recapitulation of in vivo cancerous microenvironments in vitro, in which the biological behaviours of cancer cells can be assessed under different metastatic conditions. Therefore, modelling bone metastasis microenvironments with 3D cultures is imperative for advancing cancer research and anti-cancer treatment strategies. In this review, multicellular tumour spheroids and bioreactors, tissue engineering constructs and scaffolds, microfluidic systems and 3D bioprinting technology are discussed to explore the progression of the 3D engineering approaches used to model the three microenvironments of bone metastasis. We aim to provide new insights into cancer biology and advance the translation of new therapies for bone metastasis.
Collapse
Affiliation(s)
- Han Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| |
Collapse
|
1029
|
Bicho D, Pina S, Oliveira JM, Reis RL. In Vitro Mimetic Models for the Bone-Cartilage Interface Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:373-394. [PMID: 29736583 DOI: 10.1007/978-3-319-76735-2_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In embryonic development, pure cartilage structures are in the basis of bone-cartilage interfaces. Despite this fact, the mature bone and cartilage structures can vary greatly in composition and function. Nevertheless, they collaborate in the osteochondral region to create a smooth transition zone that supports the movements and forces resulting from the daily activities. In this sense, all the hierarchical organization is involved in the maintenance and reestablishment of the equilibrium in case of damage. Therefore, this interface has attracted a great deal of interest in order to understand the mechanisms of regeneration or disease progression in osteoarthritis. With that purpose, in vitro tissue models (either static or dynamic) have been studied. Static in vitro tissue models include monocultures, co-cultures, 3D cultures, and ex vivo cultures, mostly cultivated in flat surfaces, while dynamic models involve the use of bioreactors and microfluidic systems. The latter have emerged as alternatives to study the cellular interactions in a more authentic manner over some disadvantages of the static models. The current alternatives of in vitro mimetic models for bone-cartilage interface regeneration are overviewed and discussed herein.
Collapse
Affiliation(s)
- Diana Bicho
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Sandra Pina
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - J Miguel Oliveira
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| |
Collapse
|
1030
|
Bidarra SJ, Barrias CC. 3D Culture of Mesenchymal Stem Cells in Alginate Hydrogels. Methods Mol Biol 2018; 2002:165-180. [PMID: 30244438 DOI: 10.1007/7651_2018_185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Three-dimensional (3D) cell culture systems have gained increasing interest among the scientific community, as they are more biologically relevant than traditional two-dimensional (2D) monolayer cultures. Alginate hydrogels can be formed under cytocompatibility conditions, being among the most widely used cell-entrapment 3D matrices. They recapitulate key structural features of the natural extracellular matrix and can be bio-functionalized with bioactive moieties, such as peptides, to specifically modulate cell behavior. Moreover, alginate viscoelastic properties can be tuned to match those of different types of native tissues. Ionic alginate hydrogels are transparent, allowing routine monitoring of entrapped cells, and crosslinking can be reverted using chelating agents for easy cell recovery. In this chapter, we describe some key steps to establish and characterize 3D cultures of mesenchymal stem cells using alginate hydrogels.
Collapse
Affiliation(s)
- Sílvia J Bidarra
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal. .,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.
| |
Collapse
|
1031
|
Dong R, Zhao X, Guo B, Ma PX. Biocompatible Elastic Conductive Films Significantly Enhanced Myogenic Differentiation of Myoblast for Skeletal Muscle Regeneration. Biomacromolecules 2017; 18:2808-2819. [DOI: 10.1021/acs.biomac.7b00749] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Ruonan Dong
- Frontier
Institute of Science and Technology, and State Key Laboratory for
Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an, 710049, China
| | - Xin Zhao
- Frontier
Institute of Science and Technology, and State Key Laboratory for
Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an, 710049, China
| | - Baolin Guo
- Frontier
Institute of Science and Technology, and State Key Laboratory for
Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an, 710049, China
| | - Peter X. Ma
- Frontier
Institute of Science and Technology, and State Key Laboratory for
Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an, 710049, China
| |
Collapse
|
1032
|
Sieck GC. Physiology in Perspective: Structure and Function-Anatomy and Physiology Are Integral. Physiology (Bethesda) 2017; 32:264-265. [PMID: 28615310 DOI: 10.1152/physiol.00014.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 11/22/2022] Open
|