1
|
Petzer M, Fobian S, Gulumian M, Steenkamp V, Cordier W. A549 Alveolar Carcinoma Spheroids as a Cytotoxicity Platform for Carboxyl- and Amine-Polyethylene Glycol Gold Nanoparticles. Pharmacol Res Perspect 2025; 13:e70051. [PMID: 39725610 PMCID: PMC11671224 DOI: 10.1002/prp2.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/13/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Gold nanoparticles (AuNPs) present with unique physicochemical features and potential for functionalization as anticancer agents. Three-dimensional spheroid models can be used to afford greater tissue representation due to their heterogeneous phenotype and complex molecular architecture. This study developed an A549 alveolar carcinoma spheroid model for cytotoxicity assessment and mechanistic evaluation of functionalized AuNPs. A549 spheroids were generated using an agarose micro-mold and were characterized (morphology, acid phosphatase activity, protein content) over 21 culturing days. The 72-h cytotoxicity of carboxyl-polyethylene glycol- (PCOOH-) and amine-polyethylene glycol- (PNH2-) functionalized AuNPs against Day 7 spheroids was assessed by determining spheroid morphology, acid phosphatase activity, protein content, caspase-3/7 activity, and cell cycle kinetics. Spheroids remained stable over the experimental period. Although the A549 spheroids' volume increased while remaining viable over the culturing period, structural integrity decreased from Day 14 onwards. The PCOOH-AuNPs lacked cytotoxicity at a maximum concentration of 1.2 × 1012 nanoparticles/mL with no prominent alteration to the cellular processes investigated, while the PNH2-AuNPs (at a maximum of 4.5 × 1012 nanoparticles/mL) displayed dose- and time-dependent cytotoxicity with associated loss of spheroid compactness, debris formation, DNA fragmentation, and a 75% reduction in acid phosphatase activity. Differentiation between cytotoxic and non-cytotoxic AuNPs was achieved, with preliminary elucidation of cytotoxicity endpoints. The PNH2-AuNPs promote cytotoxicity by modulating cellular kinetics while destabilizing the spheroid ultrastructure. The model serves as a proficient platform for more in-depth elucidation of NP cytotoxicity at the preclinical investigation phase.
Collapse
Affiliation(s)
- Melissa Petzer
- Department of Pharmacology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Seth‐Frerich Fobian
- Department of Pharmacology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Mary Gulumian
- Molecular Medicine and Haematology, School of PathologyUniversity of WitwatersrandJohannesburgSouth Africa
- Water Research Group, Unit for Environmental Sciences and ManagementNorth‐West UniversityPotchefstroomSouth Africa
| | - Vanessa Steenkamp
- Department of Pharmacology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Werner Cordier
- Department of Pharmacology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| |
Collapse
|
2
|
Yang S, Leung AYP, Wang Z, Yiu CKY, Dissanayaka WL. Proanthocyanidin surface preconditioning of dental pulp stem cell spheroids enhances dimensional stability and biomineralization in vitro. Int Endod J 2024; 57:1639-1654. [PMID: 39046812 DOI: 10.1111/iej.14126] [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: 11/27/2023] [Revised: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
AIM Lack of adequate mechanical strength and progressive shrinkage over time remain challenges in scaffold-free microtissue-based dental pulp regeneration. Surface collagen cross-linking holds the promise to enhance the mechanical stability of microtissue constructs and trigger biological regulations. In this study, we proposed a novel strategy for surface preconditioning microtissues using a natural collagen cross-linker, proanthocyanidin (PA). We evaluated its effects on cell viability, tissue integrity, and biomineralization of dental pulp stem cell (DPSCs)-derived 3D cell spheroids. METHODOLOGY Microtissue and macrotissue spheroids were fabricated from DPSCs and incubated with PA solution for surface collagen cross-linking. Microtissue viability was examined by live/dead staining and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, with transverse dimension change monitored. Microtissue surface stiffness was measured by an atomic force microscope (AFM). PA-preconditioned microtissues and macrotissues were cultured under basal or osteogenic conditions. Immunofluorescence staining of PA-preconditioned microtissues was performed to detect dentin sialophosphoprotein (DSPP) and F-actin expressions. PA-preconditioned macrotissues were subjected to histological analysis, including haematoxylin-eosin (HE), alizarin red, and Masson trichrome staining. Immunohistochemistry staining was used to detect alkaline phosphatase (ALP) and dentin matrix acidic phosphoprotein 1 (DMP-1) expressions. RESULTS PA preconditioning had no adverse effects on microtissue spheroid viability and increased surface stiffness. It reduced dimensional shrinkage for over 7 days in microtissues and induced a larger transverse-section area in the macrotissue. PA preconditioning enhanced collagen formation, mineralized nodule formation, and elevated ALP and DMP-1 expressions in macrotissues. Additionally, PA preconditioning induced higher F-actin and DSPP expression in microtissues, while inhibition of F-actin activity by cytochalasin B attenuated PA-induced dimensional change and DSPP upregulation. CONCLUSION PA surface preconditioning of DPSCs spheroids demonstrates excellent biocompatibility while effectively enhancing tissue structure stability and promoting biomineralization. This strategy strengthens tissue integrity in DPSC-derived spheroids and amplifies osteogenic differentiation potential, advancing scaffold-free tissue engineering applications in regenerative dentistry.
Collapse
Affiliation(s)
- Shengyan Yang
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Andy Yu Pan Leung
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Zheng Wang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry & Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Waruna Lakmal Dissanayaka
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
3
|
Wester M, Lim J, Khaertdinova L, Darsi S, Donthamsetti N, Mensing G, Vasmatzis G, Anastasiadis P, Valera E, Bashir R. On the design and fabrication of nanoliter-volume hanging drop networks. MICROSYSTEMS & NANOENGINEERING 2024; 10:147. [PMID: 39414790 PMCID: PMC11484691 DOI: 10.1038/s41378-024-00788-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 10/18/2024]
Abstract
Hanging drop cultures provide a favorable environment for the gentle, gel-free formation of highly uniform three-dimensional cell cultures often used in drug screening applications. Initial cell numbers can be limited, as with primary cells provided by minimally invasive biopsies. Therefore, it can be beneficial to divide cells into miniaturized arrays of hanging drops to supply a larger number of samples. Here, we present a framework for the miniaturization of hanging drop networks to nanoliter volumes. The principles of a single hanging drop are described and used to construct the fundamental equations for a microfluidic system composed of multiple connected drops. Constitutive equations for the hanging drop as a nonlinear capacitive element are derived for application in the electronic-hydraulic analogy, forming the basis for more complex, time-dependent numerical modeling of hanging drop networks. This is supplemented by traditional computational fluid dynamics simulation to provide further information about flow conditions within the wells. A fabrication protocol is presented and demonstrated for creating transparent, microscale arrays of pinned hanging drops. A custom interface, pressure-based fluidic system, and environmental chamber have been developed to support the device. Finally, fluid flow on the chip is demonstrated to align with expected behavior based on the principles derived for hanging drop networks. Challenges with the system and potential areas for improvement are discussed. This paper expands on the limited body of hanging drop network literature and provides a framework for designing, fabricating, and operating these systems at the microscale.
Collapse
Affiliation(s)
- Matthew Wester
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jongwon Lim
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Liliana Khaertdinova
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sriya Darsi
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Neel Donthamsetti
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Glennys Mensing
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - George Vasmatzis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, 61801, USA
| | - Panos Anastasiadis
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, 61801, USA
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Enrique Valera
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Biomedical Research Center, Carle Foundation Hospital, Urbana, IL, 61801, USA
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Mayo-Illinois Alliance for Technology-Based Healthcare, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Biomedical Research Center, Carle Foundation Hospital, Urbana, IL, 61801, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Biomedical and Translation Science, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, 60642, USA.
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
4
|
Lee S, Woo CJ, Jung HI, Nam KC, Lim JS, Kwak BS. Formation Pattern Analysis of Spheroids Formed by a Droplet-Based Microfluidic System for Predicting the Aggressiveness of Tumor Cells. ACS Biomater Sci Eng 2024; 10:2477-2485. [PMID: 38483467 DOI: 10.1021/acsbiomaterials.4c00005] [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] [Indexed: 04/09/2024]
Abstract
Examining tumor heterogeneity is essential for selecting an appropriate anticancer treatment for an individual. This study aimed to distinguish low- and high-aggressive tumor cells by analyzing the formation patterns of spheroids. The droplet-based microfluidic system was employed for the formation of each spheroid from four different subtypes of breast tumor cells. Additionally, heterotypic spheroids with T lymphocytes and cancer-associated fibroblasts (CAFs) were produced, and distinctions between low- and high-aggressive tumor cells were explored through the analysis of formation patterns using circularity, convexity, and cell distributions. In both homotypic spheroids and heterotypic spheroids with T lymphocytes, spheroids formed from low-aggressive tumor cells exhibited high circularity and convexity. On the other hand, spheroids formed from high-aggressive tumor cells had relatively low circularity and convexity. In the case of heterotypic spheroids with CAFs, circularity and convexity did not exhibit clear differences between low- and high-aggressive tumor cells, but distinct variations were observed in cell distributions. CAFs and low-aggressive tumor cells were evenly distributed, whereas the CAFs were predominantly located in the inner layer, and high-aggressive tumor cells were primarily located in the outer layer. This finding can offer valuable insights into predicting the aggressiveness of unknown tumor cells.
Collapse
Affiliation(s)
- Sunghan Lee
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi 10326, Gyeonggi-do, Republic of Korea
| | - Chang Jae Woo
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi 10326, Gyeonggi-do, Republic of Korea
- National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si 10408, Gyeonggi-do, Republic of Korea
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seadaemun-gu, Seoul 03722, Republic of Korea
- The DABOM Inc., Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ki Chang Nam
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi 10326, Gyeonggi-do, Republic of Korea
| | - Ji Seok Lim
- School of Mechanical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan-si 38541, Gyeongsanbuk-do, Republic of Korea
- MediSphere Inc., Gyeongsan-si 38541, Gyeongsanbuk-do, Republic of Korea
| | - Bong Seop Kwak
- College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyangsi 10326, Gyeonggi-do, Republic of Korea
- MediSphere Inc., Gyeongsan-si 38541, Gyeongsanbuk-do, Republic of Korea
| |
Collapse
|
5
|
Hu J, Liu K, Ghosh C, Khaket TP, Shih H, Kebebew E. Anaplastic thyroid cancer spheroids as preclinical models to test therapeutics. J Exp Clin Cancer Res 2024; 43:85. [PMID: 38500204 PMCID: PMC10949686 DOI: 10.1186/s13046-024-03009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/10/2024] [Indexed: 03/20/2024] Open
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive thyroid cancer. Despite advances in tissue culture techniques, a robust model for ATC spheroid culture is yet to be developed. In this study, we created an efficient and cost-effective 3D tumor spheroids culture system from human ATC cells and existing cell lines that better mimic patient tumors and that can enhance our understanding of in vivo treatment response. We found that patient-derived ATC cells and cell lines can readily form spheroids in culture with a unique morphology, size, and cytoskeletal organization. We observed both cohesive (dense and solid structures) and discohesive (irregularly shaped structures) spheroids within the same culture condition across different cell lines. BRAFWT ATC spheroids grew in a cohesive pattern, while BRAFV600E-mutant ATC spheroids had a discohesive organization. In the patient-derived BRAFV600E-mutant ATC spheroids, we observed both growth patterns, but mostly the discohesive type. Histologically, ATC spheroids had a similar morphology to the patient's tumor through H&E staining and proliferation marker staining. Moreover, RNA sequencing analysis revealed that the gene expression profile of tumor cells derived from the spheroids closely matched parental patient tumor-derived cells in comparison to monolayer cultures. In addition, treatment response to combined BRAF and MEK inhibition in BRAFV600E-mutant ATC spheroids exhibited a similar sensitivity to the patient clinical response. Our study provides a robust and novel ex vivo spheroid model system that can be used in both established ATC cell lines and patient-derived tumor samples to better understand the biology of ATC and to test therapeutics.
Collapse
Affiliation(s)
- Jiangnan Hu
- Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Chandrayee Ghosh
- Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Tejinder Pal Khaket
- Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Helen Shih
- Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Electron Kebebew
- Department of Surgery and Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
6
|
Kostadinova R, Ströbel S, Chen L, Fiaschetti-Egli K, Gadient J, Pawlowska A, Petitjean L, Bieri M, Thoma E, Petitjean M. Digital pathology with artificial intelligence analysis provides insight to the efficacy of anti-fibrotic compounds in human 3D MASH model. Sci Rep 2024; 14:5885. [PMID: 38467661 PMCID: PMC10928082 DOI: 10.1038/s41598-024-55438-2] [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: 11/10/2023] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a severe liver disease characterized by lipid accumulation, inflammation and fibrosis. The development of MASH therapies has been hindered by the lack of human translational models and limitations of analysis techniques for fibrosis. The MASH three-dimensional (3D) InSight™ human liver microtissue (hLiMT) model recapitulates pathophysiological features of the disease. We established an algorithm for automated phenotypic quantification of fibrosis of Sirius Red stained histology sections of MASH hLiMTs model using a digital pathology quantitative single-fiber artificial intelligence (AI) FibroNest™ image analysis platform. The FibroNest™ algorithm for MASH hLiMTs was validated using anti-fibrotic reference compounds with different therapeutic modalities-ALK5i and anti-TGF-β antibody. The phenotypic quantification of fibrosis demonstrated that both reference compounds decreased the deposition of fibrillated collagens in alignment with effects on the secretion of pro-collagen type I/III, tissue inhibitor of metalloproteinase-1 and matrix metalloproteinase-3 and pro-fibrotic gene expression. In contrast, clinical compounds, Firsocostat and Selonsertib, alone and in combination showed strong anti-fibrotic effects on the deposition of collagen fibers, however less pronounced on the secretion of pro-fibrotic biomarkers. In summary, the phenotypic quantification of fibrosis of MASH hLiMTs combined with secretion of pro-fibrotic biomarkers and transcriptomics represents a promising drug discovery tool for assessing anti-fibrotic compounds.
Collapse
Affiliation(s)
| | - Simon Ströbel
- InSphero AG, Wagistrasse 27A, Schlieren, Switzerland
| | - Li Chen
- PharmaNest, Princeton, NJ, USA
| | | | - Jana Gadient
- InSphero AG, Wagistrasse 27A, Schlieren, Switzerland
| | | | | | - Manuela Bieri
- InSphero AG, Wagistrasse 27A, Schlieren, Switzerland
| | - Eva Thoma
- InSphero AG, Wagistrasse 27A, Schlieren, Switzerland
| | | |
Collapse
|
7
|
Yang GD, Ma DS, Ma CY, Bai Y. Research Progress on Cardiac Tissue Construction of Mesenchymal Stem Cells for Myocardial Infarction. Curr Stem Cell Res Ther 2024; 19:942-958. [PMID: 37612870 DOI: 10.2174/1574888x18666230823091017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023]
Abstract
Heart failure is still the main complication affecting the prognosis of acute myocardial infarction (AMI), and mesenchymal stem cells (MSCs) are an effective treatment to replace necrotic myocardium and improve cardiac functioning. However, the transplant survival rate of MSCs still presents challenges. In this review, the biological characteristics of MSCs, the progress of mechanism research in the treatment of myocardial infarction, and the advances in improving the transplant survival rate of MSCs in the replacement of necrotic myocardial infarction are systematically described. From a basic to advanced clinical research, MSC transplants have evolved from a pure injection, an exosome injection, the genetic modification of MSCs prior to injection to the cardiac tissue engineering of MSC patch grafting. This study shows that MSCs have wide clinical applications in the treatment of AMI, suggesting improved myocardial tissue creation. A broader clinical application prospect will be explored and developed to improve the survival rate of MSC transplants and myocardial vascularization.
Collapse
Affiliation(s)
- Guo-Dong Yang
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Da-Shi Ma
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chun-Ye Ma
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yang Bai
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
8
|
Arutyunyan I, Jumaniyazova E, Makarov A, Fatkhudinov T. In Vitro Models of Head and Neck Cancer: From Primitive to Most Advanced. J Pers Med 2023; 13:1575. [PMID: 38003890 PMCID: PMC10672510 DOI: 10.3390/jpm13111575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
For several decades now, researchers have been trying to answer the demand of clinical oncologists to create an ideal preclinical model of head and neck squamous cell carcinoma (HNSCC) that is accessible, reproducible, and relevant. Over the past years, the development of cellular technologies has naturally allowed us to move from primitive short-lived primary 2D cell cultures to complex patient-derived 3D models that reproduce the cellular composition, architecture, mutational, or viral load of native tumor tissue. Depending on the tasks and capabilities, a scientific laboratory can choose from several types of models: primary cell cultures, immortalized cell lines, spheroids or heterospheroids, tissue engineering models, bioprinted models, organoids, tumor explants, and histocultures. HNSCC in vitro models make it possible to screen agents with potential antitumor activity, study the contribution of the tumor microenvironment to its progression and metastasis, determine the prognostic significance of individual biomarkers (including using genetic engineering methods), study the effect of viral infection on the pathogenesis of the disease, and adjust treatment tactics for a specific patient or groups of patients. Promising experimental results have created a scientific basis for the registration of several clinical studies using HNSCC in vitro models.
Collapse
Affiliation(s)
- Irina Arutyunyan
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov Ministry of Healthcare of the Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
| | - Andrey Makarov
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- Histology Department, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 117997 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (I.A.); (A.M.); (T.F.)
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
9
|
Tabatabaei Rezaei N, Kumar H, Liu H, Lee SS, Park SS, Kim K. Recent Advances in Organ-on-Chips Integrated with Bioprinting Technologies for Drug Screening. Adv Healthc Mater 2023; 12:e2203172. [PMID: 36971091 PMCID: PMC11469032 DOI: 10.1002/adhm.202203172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Currently, the demand for more reliable drug screening devices has made scientists and researchers develop novel potential approaches to offer an alternative to animal studies. Organ-on-chips are newly emerged platforms for drug screening and disease metabolism investigation. These microfluidic devices attempt to recapitulate the physiological and biological properties of different organs and tissues using human-derived cells. Recently, the synergistic combination of additive manufacturing and microfluidics has shown a promising impact on improving a wide array of biological models. In this review, different methods are classified using bioprinting to achieve the relevant biomimetic models in organ-on-chips, boosting the efficiency of these devices to produce more reliable data for drug investigations. In addition to the tissue models, the influence of additive manufacturing on microfluidic chip fabrication is discussed, and their biomedical applications are reviewed.
Collapse
Affiliation(s)
- Nima Tabatabaei Rezaei
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Hitendra Kumar
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
- Department of Pathology and Laboratory MedicineCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Hongqun Liu
- Liver UnitCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Samuel S. Lee
- Liver UnitCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Simon S. Park
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
- Department of Biomedical EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| |
Collapse
|
10
|
Carannante V, Wiklund M, Önfelt B. In vitro models to study natural killer cell dynamics in the tumor microenvironment. Front Immunol 2023; 14:1135148. [PMID: 37457703 PMCID: PMC10338882 DOI: 10.3389/fimmu.2023.1135148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Immunotherapy is revolutionizing cancer therapy. The rapid development of new immunotherapeutic strategies to treat solid tumors is posing new challenges for preclinical research, demanding novel in vitro methods to test treatments. Such methods should meet specific requirements, such as enabling the evaluation of immune cell responses like cytotoxicity or cytokine release, and infiltration into the tumor microenvironment using cancer models representative of the original disease. They should allow high-throughput and high-content analysis, to evaluate the efficacy of treatments and understand immune-evasion processes to facilitate development of new therapeutic targets. Ideally, they should be suitable for personalized immunotherapy testing, providing information for patient stratification. Consequently, the application of in vitro 3-dimensional (3D) cell culture models, such as tumor spheroids and organoids, is rapidly expanding in the immunotherapeutic field, coupled with the development of novel imaging-based techniques and -omic analysis. In this paper, we review the recent advances in the development of in vitro 3D platforms applied to natural killer (NK) cell-based cancer immunotherapy studies, highlighting the benefits and limitations of the current methods, and discuss new concepts and future directions of the field.
Collapse
Affiliation(s)
- Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Wiklund
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Goel R, Gulwani D, Upadhyay P, Sarangthem V, Singh TD. Unsung versatility of elastin-like polypeptide inspired spheroid fabrication: A review. Int J Biol Macromol 2023; 234:123664. [PMID: 36791934 DOI: 10.1016/j.ijbiomac.2023.123664] [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: 12/03/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Lately, 3D cell culture technique has gained a lot of appreciation as a research model. Augmented with technological advancements, the area of 3D cell culture is growing rapidly with a diverse array of scaffolds being tested. This is especially the case for spheroid cultures. The culture of cells as spheroids provides opportunities for unanticipated vision into biological phenomena with its application to drug discovery, metabolic profiling, stem cell research as well as tumor, and disease biology. Spheroid fabrication techniques are broadly categorised into matrix-dependent and matrix-independent techniques. While there is a profusion of spheroid fabrication substrates with substantial biological relevance, an economical, modular, and bio-compatible substrate for high throughput production of spheroids is lacking. In this review, we posit the prospects of elastin-like polypeptides (ELPs) as a broad-spectrum spheroid fabrication platform. Elastin-like polypeptides are nature inspired, size-tunable genetically engineered polymers with wide applicability in various arena of biological considerations, has been employed for spheroid culture with profound utility. The technology offers a cheap, high-throughput, reproducible alternative for spheroid culture with exquisite adaptability. Here, we will brief the applicability of 3D cultures as compared to 2D cultures with spheroids being the focal point of the review. Common approaches to spheroid fabrication are discussed with existential limitations. Finally, the versatility of elastin-like polypeptide inspired substrates for spheroid culture has been discussed.
Collapse
Affiliation(s)
- Ridhima Goel
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Deepak Gulwani
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Priyanka Upadhyay
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Sarangthem
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
12
|
Otsuka H. Nanofabrication Technologies to Control Cell and Tissue Function in Three-Dimension. Gels 2023; 9:gels9030203. [PMID: 36975652 PMCID: PMC10048556 DOI: 10.3390/gels9030203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/29/2023] Open
Abstract
In the 2000s, advances in cellular micropatterning using microfabrication contributed to the development of cell-based biosensors for the functional evaluation of newly synthesized drugs, resulting in a revolutionary evolution in drug screening. To this end, it is essential to utilize cell patterning to control the morphology of adherent cells and to understand contact and paracrine-mediated interactions between heterogeneous cells. This suggests that the regulation of the cellular environment by means of microfabricated synthetic surfaces is not only a valuable endeavor for basic research in biology and histology, but is also highly useful to engineer artificial cell scaffolds for tissue regeneration. This review particularly focuses on surface engineering techniques for the cellular micropatterning of three-dimensional (3D) spheroids. To establish cell microarrays, composed of a cell adhesive region surrounded by a cell non-adherent surface, it is quite important to control a protein-repellent surface in the micro-scale. Thus, this review is focused on the surface chemistries of the biologically inspired micropatterning of two-dimensional non-fouling characters. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single-cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., fibers and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. These important approaches to cell engineering result in their applications to tissue regeneration, where the cell-biomaterial composite is injected into diseased area. This approach allows the operating surgeon to implant the cell and polymer combinations with minimum invasiveness. The polymers utilized in hydrogels are structurally similar to components of the extracellular matrix in vivo, and are considered biocompatible. This review will provide an overview of the critical design to make hydrogels when used as cell scaffolds for tissue engineering. In addition, the new strategy of injectable hydrogel will be discussed as future directions.
Collapse
Affiliation(s)
- Hidenori Otsuka
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, 1-3 Kagurazaka, Shinjuku-ku, Tokyo 162-8601, Japan
| |
Collapse
|
13
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Beatriz AL, Nolwenn H. Innovative in vitro approaches to toxicological investigations of mycotoxins effects. EFSA J 2022; 20:e200907. [PMID: 36531276 PMCID: PMC9749439 DOI: 10.2903/j.efsa.2022.e200907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Among the potential contaminants, mycotoxins are of particular concern due to the importance in terms of food and feed safety. The difficulty in establishing a diagnosis for mycotoxicosis relies in the fact that the effects are subclinical, and that multicontamination by various toxins is the most common scenario. The co-occurrence of these mycotoxins raises questions concerning both food safety and regulation. However, there is still limited knowledge on toxicity data on co-exposure. The current technical report will describe the activities performed by the fellow in the LUBEM-Brest University (France). In this context, the work programme offered by the hosting site consisted in vitro toxicological approaches to evaluate the toxicity of mycotoxin mixtures. The aim of this project was to assess human risk to the exposure of two main regulated mycotoxins (ochratoxin A and fumonisin B1) using different innovative cellular models (2D and 3D spheroids). In this framework, these mycotoxins were tested individually and as a combination on intestinal and hepatic cell lines alone or in co-cultures. Overall, our results show the outstanding potential of using more predictive and realistic approaches for the risk assessment (RA) of mycotoxins. It is of high importance to pursue further toxicological characterisations and exposure evaluations for mycotoxins, in order to determine a more detailed RA. This will serve as a reference to understand multicontamination mechanism of mycotoxins at the cell level and help authority to revise regulation.
Collapse
Affiliation(s)
- Arce López Beatriz
- Université de Bretagne Occidentale ‐ (Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, LUBEM) (UBO)France
| | - Hymery Nolwenn
- Université de Bretagne Occidentale ‐ (Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, LUBEM) (UBO)France
| |
Collapse
|
15
|
3D Bioprinting of Smart Oxygen-Releasing Cartilage Scaffolds. J Funct Biomater 2022; 13:jfb13040252. [PMID: 36412893 PMCID: PMC9680294 DOI: 10.3390/jfb13040252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Three-dimensional bioprinting is a powerful technique for manufacturing improved engineered tissues. Three-dimensional bioprinted hydrogels have significantly advanced the medical field to repair cartilage tissue, allowing for such constructs to be loaded with different components, such as cells, nanoparticles, and/or drugs. Cartilage, as an avascular tissue, presents extreme difficulty in self-repair when it has been damaged. In this way, hydrogels with optimal chemical and physical properties have been researched to respond to external stimuli and release various bioactive agents to further promote a desired tissue response. For instance, methacryloyl gelatin (GelMA) is a type of modified hydrogel that allows for the encapsulation of cells, as well as oxygen-releasing nanoparticles that, in the presence of an aqueous medium and through controlled porosity and swelling, allow for internal and external environmental exchanges. This review explores the 3D bioprinting of hydrogels, with a particular focus on GelMA hydrogels, to repair cartilage tissue. Recent advances and future perspectives are described.
Collapse
|
16
|
Kim DE, Lee YB, Shim HE, Song JJ, Han JS, Moon KS, Huh KM, Kang SW. Application of Hexanoyl Glycol Chitosan as a Non-cell Adhesive Polymer in Three-Dimensional Cell Culture. ACS OMEGA 2022; 7:18471-18480. [PMID: 35694497 PMCID: PMC9178711 DOI: 10.1021/acsomega.2c00890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Cell culture technology has evolved into three-dimensional (3D) artificial tissue models for better reproduction of human native tissues. However, there are some unresolved limitations that arise due to the adhesive properties of cells. In this study, we developed a hexanoyl glycol chitosan (HGC) as a non-cell adhesive polymer for scaffold-based and -free 3D culture. The uniform cell distribution in a porous scaffold was well maintained during the long culutre period on the HGC-coated substrate by preventing ectopic adhesion and migration of cells on the substrate. In addition, when culturing many spheroids in one dish, supplementation of the culture medium with HGC prevented the aggregation of spheroids and maintained the shape and size of spheroids for a long culture duration. Collectively, the use of HGC in 3D culture systems is expected to contribute greatly to creating excellent regenerative therapeutics and screening models of bioproducts.
Collapse
Affiliation(s)
- Da-Eun Kim
- Research
Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department
of Polymer Science and Engineering, Chungnam
National University, Daejeon 34134, Republic of Korea
| | - Yu Bin Lee
- Department
of Advanced Toxicology Research, Korea Institute
of Toxicology, Daejeon 34114, Republic of Korea
| | - Hye-Eun Shim
- Research
Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department
of Polymer Science and Engineering, Chungnam
National University, Daejeon 34134, Republic of Korea
| | - Jin Jung Song
- Research
Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Department
of Polymer Science and Engineering, Chungnam
National University, Daejeon 34134, Republic of Korea
| | - Ji-Seok Han
- Department
of Toxicological Evaluation and Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kyoung-Sik Moon
- Department
of Advanced Toxicology Research, Korea Institute
of Toxicology, Daejeon 34114, Republic of Korea
| | - Kang Moo Huh
- Department
of Polymer Science and Engineering, Chungnam
National University, Daejeon 34134, Republic of Korea
| | - Sun-Woong Kang
- Research
Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
- Human
and Environmental Toxicology Program, University
of Science and Technology, Daejeon 34114, Republic of Korea
| |
Collapse
|
17
|
Ströbel S, Kostadinova R, Fiaschetti-Egli K, Rupp J, Bieri M, Pawlowska A, Busler D, Hofstetter T, Sanchez K, Grepper S, Thoma E. A 3D primary human cell-based in vitro model of non-alcoholic steatohepatitis for efficacy testing of clinical drug candidates. Sci Rep 2021; 11:22765. [PMID: 34815444 PMCID: PMC8611054 DOI: 10.1038/s41598-021-01951-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive and severe liver disease, characterized by lipid accumulation, inflammation, and downstream fibrosis. Despite its increasing prevalence, there is no approved treatment yet available for patients. This has been at least partially due to the lack of predictive preclinical models for studying this complex disease. Here, we present a 3D in vitro microtissue model that uses spheroidal, scaffold free co-culture of primary human hepatocytes, Kupffer cells, liver endothelial cells and hepatic stellate cells. Upon exposure to defined and clinically relevant lipotoxic and inflammatory stimuli, these microtissues develop key pathophysiological features of NASH within 10 days, including an increase of intracellular triglyceride content and lipids, and release of pro-inflammatory cytokines. Furthermore, fibrosis was evident through release of procollagen type I, and increased deposition of extracellular collagen fibers. Whole transcriptome analysis revealed changes in the regulation of pathways associated with NASH, such as lipid metabolism, inflammation and collagen processing. Importantly, treatment with anti-NASH drug candidates (Selonsertib and Firsocostat) decreased the measured specific disease parameter, in accordance with clinical observations. These drug treatments also significantly changed the gene expression patterns of the microtissues, thus providing mechanisms of action and revealing therapeutic potential. In summary, this human NASH model represents a promising drug discovery tool for understanding the underlying complex mechanisms in NASH, evaluating efficacy of anti-NASH drug candidates and identifying new approaches for therapeutic interventions.
Collapse
Affiliation(s)
- Simon Ströbel
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH, Switzerland.
| | | | | | - Jana Rupp
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | - Manuela Bieri
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | | | - Donna Busler
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | | | | | - Sue Grepper
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| | - Eva Thoma
- InSphero AG, Wagistrasse 27A, 8952 Schlieren, CH Switzerland
| |
Collapse
|
18
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
19
|
Lewis-Israeli YR, Wasserman AH, Aguirre A. Heart Organoids and Engineered Heart Tissues: Novel Tools for Modeling Human Cardiac Biology and Disease. Biomolecules 2021; 11:1277. [PMID: 34572490 PMCID: PMC8468189 DOI: 10.3390/biom11091277] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/06/2021] [Accepted: 08/24/2021] [Indexed: 01/02/2023] Open
Abstract
Organoids are three-dimensional in vitro cell constructs that recapitulate organ properties and structure to a significant extent. They constitute particularly useful models to study unapproachable states in humans, such as embryonic and fetal development, or early disease progression in adults. In recent years organoids have been implemented to model a wide range of different organs and disease conditions. However, the technology for their fabrication and application to cardiovascular studies has been lagging significantly when compared to other organoid types (e.g., brain, pancreas, kidney, intestine). This is a surprising fact since cardiovascular disease (CVD) and congenital heart disease (CHD) constitute the leading cause of mortality and morbidity in the developed world, and the most common birth defect in humans, respectively, and collectively constitute one of the largest unmet medical needs in the modern world. There is a critical need to establish in vitro models of the human heart that faithfully recapitulate its biology and function, thus enabling basic and translational studies to develop new therapeutics. Generating heart organoids that truly resemble the heart has proven difficult due to its complexity, but significant progress has been made recently to overcome this obstacle. In this review, we will discuss progress in novel heart organoid generation methods, the advantages and disadvantages of each approach, and their translational applications for advancing cardiovascular studies and the treatment of heart disorders.
Collapse
Affiliation(s)
- Yonatan R. Lewis-Israeli
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA; (Y.R.L.-I.); (A.H.W.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Aaron H. Wasserman
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA; (Y.R.L.-I.); (A.H.W.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Aitor Aguirre
- Division of Developmental and Stem Cell Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA; (Y.R.L.-I.); (A.H.W.)
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48823, USA
| |
Collapse
|
20
|
Chu X, Wang M, Qiu X, Huang Y, Li T, Otieno E, Li N, Luo L, Xiao X. Strategies for constructing pluripotent stem cell- and progenitor cell-derived three-dimensional cardiac micro-tissues. J Biomed Mater Res A 2021; 110:488-503. [PMID: 34397148 DOI: 10.1002/jbm.a.37298] [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: 02/20/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cardiac micro-tissue is a promising model for simulating the structural and functional features of heart in vitro. This scientific achievement provides a platform for exploration about the mechanisms on the development, damage, and regeneration of tissue, hence, paving a way toward development of novel therapies for heart diseases. However, 3D micro-tissue technology is still in its infant stages faced with many challenges such as incompleteness of the tissue microarchitecture, loss of the resident immune cells, poor reproducibility, and deficiencies in continuously feeding the nutrients and removing wastes during micro-tissue culturing. There is an urgent need to optimize the construction of 3D cardiac micro-tissue and improve functions of the involved cells. Therefore, scaffolds and cell resources for building 3D cardiac micro-tissues, strategies for inducing the maturation and functionalization of pluripotent stem cell- or cardiac progenitor cell-derived cardiomyocytes, and the major challenges were reviewed in this writing to enable future fabrication of 3D cardiac micro-tissues or organoids for drug screening, disease modeling, regeneration treatment, and so on.
Collapse
Affiliation(s)
- Xinyue Chu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mingyu Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China.,Institute of Laboratory Animals Science, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoyan Qiu
- Department of Animal Husbandry Engineering, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yun Huang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Tong Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Edward Otieno
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Na Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Li Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiong Xiao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
21
|
Zhang J, Xu W, Li C, Meng F, Guan Y, Liu X, Zhao J, Peng J, Wang Y. Tissue Engineering Microtissue: Construction, Optimization, and Application. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:393-404. [PMID: 33719547 DOI: 10.1089/ten.teb.2020.0370] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Until now, there is no clear definition of microtissue; it usually refers to the microtissue formed by the aggregation of seed cells under the action of cell-cell or cell-extracellular matrix (ECM). Compared with traditional cell monolayer culture, cells are cultivated into a three-dimensional microstructure in a specific way. The microstructure characteristics of microtissue are similar to natural tissues and can promote cell proliferation and differentiation. Therefore, it has a broader range of biomedical applications in tissue engineering. The traditional tissue engineering strategy is to add high-density seed cells and biomolecules on a preformed scaffold to construct a tissue engineering graft. However, due to the destruction of the ECM of the cells cultured in a monolayer during the digestion process with trypsin, the uneven distribution of the cells in the scaffold, and the damage of various adverse factors after the cells are implanted in the scaffold, this strategy is often ineffective, and the subsequent applications still face challenges. This article reviews the latest researches of a new strategy-tissue engineering microtissue strategy; discuss several traditional construction methods, structure, and function optimization; and practical application of microtissue. The review aims to provide a reference for future research on tissue engineering microtissue. Impact statement The traditional tissue engineering strategies have several disadvantages, researchers have conducted extensive research on tissue engineering microtissues in recent years, and they make significant progress. Microtissue is a kind of microtissue with three-dimensional structure, its microstructure is similar to that of natural tissue. In addition, microtissue implantation can protect cells from mechanical interference, inflammation, and other adverse factors. Furthermore, it improves the survival rate of cells and the therapeutic effect of tissue-engineered grafts. However, the practical conditions, advantages, and disadvantages of tissue engineering microtissue have not been fully elucidated. The purpose of this review is to discuss the latest research progress of microtissue and provide a reference for future research.
Collapse
Affiliation(s)
- Jian Zhang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Wenjing Xu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Chaochao Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Fanqi Meng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Department of Spine Surgery, Peking University People's Hospital, Beijing, P.R. China
| | - Yanjun Guan
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiuzhi Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Jie Zhao
- Beijing Tsinghua Changgeng Hospital Affiliated to Tsinghua University, Tsinghua University Clinical School, Beijing, P.R. China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China
| |
Collapse
|
22
|
Sakalem ME, De Sibio MT, da Costa FADS, de Oliveira M. Historical evolution of spheroids and organoids, and possibilities of use in life sciences and medicine. Biotechnol J 2021; 16:e2000463. [PMID: 33491924 DOI: 10.1002/biot.202000463] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND An impressive percentage of biomedical advances were achieved through animal research and cell culture investigations. For drug testing and disease researches, both animal models and preclinical trials with cell cultures are extremely important, but present some limitations, such as ethical concern and inability of representing complex tissues and organs. 3D cell cultures arise providing a more realistic in vitro representation of tissues and organs. Environment and cell type in 3D cultures can represent in vivo conditions and thus provide accurate data on cell-to-cell interactions, and cultivation techniques are based on a scaffold, usually hydrogel or another polymeric material, or without scaffold, such as suspended microplates, magnetic levitation, and microplates for spheroids with ultra-low fixation coating. PURPOSE AND SCOPE This review aims at presenting an updated summary of the most common 3D cell culture models available, as well as a historical background of their establishment and possible applications. SUMMARY Even though 3D culturing is incapable of replacing other current research types, they will continue to substitute some unnecessary animal experimentation, as well as complement monolayer cultures. CONCLUSION In this aspect, 3D culture emerges as a valuable alternative to the investigation of functional, biochemical, and molecular aspects of human pathologies.
Collapse
Affiliation(s)
| | - Maria Teresa De Sibio
- Department of Internal Clinic, Botucatu Medicine School of the Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Felipe Allan da Silva da Costa
- Department of Bioprocesses and Biotechnology, School of Agricultural Sciences of the Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Miriane de Oliveira
- Department of Internal Clinic, Botucatu Medicine School of the Sao Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
23
|
Generation of immune cell containing adipose organoids for in vitro analysis of immune metabolism. Sci Rep 2020; 10:21104. [PMID: 33273595 PMCID: PMC7713299 DOI: 10.1038/s41598-020-78015-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is an organized endocrine organ with important metabolic and immunological functions and immune cell-adipocyte crosstalk is known to drive various disease pathologies. Suitable 3D adipose tissue organoid models often lack resident immune cell populations and therefore require the addition of immune cells isolated from other organs. We have created the first 3D adipose tissue organoid model which could contain and maintain resident immune cell populations of the stromal vascular fraction (SVF) and proved to be effective in studying adipose tissue biology in a convenient manner. Macrophage and mast cell populations were successfully confirmed within our organoid model and were maintained in culture without the addition of growth factors. We demonstrated the suitability of our model for monitoring the lipidome during adipocyte differentiation in vitro and confirmed that this model reflects the physiological lipidome better than standard 2D cultures. In addition, we applied mass spectrometry-based lipidomics to track lipidomic changes in the lipidome upon dietary and immunomodulatory interventions. We conclude that this model represents a valuable tool for immune-metabolic research.
Collapse
|
24
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
25
|
Dissanayaka WL, Zhang C. Scaffold-based and Scaffold-free Strategies in Dental Pulp Regeneration. J Endod 2020; 46:S81-S89. [DOI: 10.1016/j.joen.2020.06.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Chen J, Huang D, Wang L, Hou J, Zhang H, Li Y, Zhong S, Wang Y, Wu Y, Huang W. 3D bioprinted multiscale composite scaffolds based on gelatin methacryloyl (GelMA)/chitosan microspheres as a modular bioink for enhancing 3D neurite outgrowth and elongation. J Colloid Interface Sci 2020; 574:162-173. [DOI: 10.1016/j.jcis.2020.04.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
|
27
|
Effect of Integrin Binding Peptide on Vascularization of Scaffold-Free Microtissue Spheroids. Tissue Eng Regen Med 2020; 17:595-605. [PMID: 32710228 DOI: 10.1007/s13770-020-00281-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/21/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Three-dimensional (3D) biomimetic models via various approaches can be used by therapeutic applications of tissue engineering. Creating an optimal vascular microenvironment in 3D model that mimics the extracellular matrix (ECM) and providing an adequate blood supply for the survival of cell transplants are major challenge that need to be overcome in tissue regeneration. However, currently available scaffolds-depended approaches fail to mimic essential functions of natural ECM. Scaffold-free microtissues (SFMs) can successfully overcome some of the major challenges caused by scaffold biomaterials such as low cell viability and high cost. METHODS Herein, we investigated the effect of soluble integrin binding peptide of arginine-glycine-aspartic acid (RGD) on vascularization of SFM spheroids of human umbilical vein endothelial cells. In vitro-fabricated microtissue spheroids were constructed and cultivated in 0 mM, 1 mM, 2 mM, and 4 mM of RGD peptide. The dimensions and viability of SFMs were measured. RESULTS Maximum dimension and cell viability observed in 2 mM RGD containing SFM. Vascular gene expression of 2 mM RGD containing SFM were higher than other groups, while 4 mM RGD containing SFM expressed minimum vascularization related genes. Immunofluorescent staining results indicating that platelet/endothelial cell adhesion molecule and vascular endothelial growth factor protein expression of 2 mM RGD containing SFM was higher compared to other groups. CONCLUSION Collectively, these findings demonstrate that SFM spheroids can be successfully vascularized in determined concentration of RGD peptide containing media. Also, soluble RGD incorporated SFMs can be used as an optimal environment for successful prevascularization strategies.
Collapse
|
28
|
Wang T, Wang L, Wang G, Zhuang Y. Leveraging and manufacturing in vitro multicellular spheroid-based tumor cell model as a preclinical tool for translating dysregulated tumor metabolism into clinical targets and biomarkers. BIORESOUR BIOPROCESS 2020. [DOI: 10.1186/s40643-020-00325-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
29
|
Wardwell-Swanson J, Suzuki M, Dowell KG, Bieri M, Thoma EC, Agarkova I, Chiovaro F, Strebel S, Buschmann N, Greve F, Frey O. A Framework for Optimizing High-Content Imaging of 3D Models for Drug Discovery. SLAS DISCOVERY 2020; 25:709-722. [PMID: 32484408 DOI: 10.1177/2472555220929291] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) spheroid models are rapidly gaining favor for drug discovery applications due to their improved morphological characteristics, cellular complexity, long lifespan in culture, and higher physiological relevance relative to two-dimensional (2D) cell culture models. High-content imaging (HCI) of 3D spheroid models has the potential to provide valuable information to help researchers untangle disease pathophysiology and assess novel therapies more effectively. The transition from 2D monolayer models to dense 3D spheroids in HCI applications is not trivial, however, and requires 3D-optimized protocols, instrumentation, and resources. Here, we discuss considerations for moving from 2D to 3D models and present a framework for HCI and analysis of 3D spheroid models in a drug discovery setting. We combined scaffold-free, multicellular spheroid models with scalable, automation-compatible plate technology enabling image-based applications ranging from high-throughput screening to more complex, lower-throughput microphysiological systems of organ networks. We used this framework in three case studies: investigation of lipid droplet accumulation in a human liver nonalcoholic steatohepatitis (NASH) model, real-time immune cell interactions in a multicellular 3D lung cancer model, and a high-throughput screening application using a 3D co-culture model of gastric carcinoma to assess dose-dependent drug efficacy and specificity. The results of these proof-of-concept studies demonstrate the potential for high-resolution image-based analysis of 3D spheroid models for drug discovery applications, and confirm that cell-level and temporal-spatial analyses that fully exploit multicellular features of spheroid models are not only possible but soon will be routine practice in drug discovery workflows.
Collapse
Affiliation(s)
| | - Mahomi Suzuki
- Yokogawa Electric Corporation, Musashino, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Verma S, Senger S, Cherayil BJ, Faherty CS. Spheres of Influence: Insights into Salmonella Pathogenesis from Intestinal Organoids. Microorganisms 2020; 8:microorganisms8040504. [PMID: 32244707 PMCID: PMC7232497 DOI: 10.3390/microorganisms8040504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/19/2022] Open
Abstract
The molecular complexity of host-pathogen interactions remains poorly understood in many infectious diseases, particularly in humans due to the limited availability of reliable and specific experimental models. To bridge the gap between classical two-dimensional culture systems, which often involve transformed cell lines that may not have all the physiologic properties of primary cells, and in vivo animal studies, researchers have developed the organoid model system. Organoids are complex three-dimensional structures that are generated in vitro from primary cells and can recapitulate key in vivo properties of an organ such as structural organization, multicellularity, and function. In this review, we discuss how organoids have been deployed in exploring Salmonella infection in mice and humans. In addition, we summarize the recent advancements that hold promise to elevate our understanding of the interactions and crosstalk between multiple cell types and the microbiota with Salmonella. These models have the potential for improving clinical outcomes and future prophylactic and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Smriti Verma
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
- Correspondence: ; Tel.: +1-617-726-7991
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| | - Bobby J. Cherayil
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| |
Collapse
|
31
|
Zhao L, Xiu J, Liu Y, Zhang T, Pan W, Zheng X, Zhang X. A 3D Printed Hanging Drop Dripper for Tumor Spheroids Analysis Without Recovery. Sci Rep 2019; 9:19717. [PMID: 31873199 PMCID: PMC6928160 DOI: 10.1038/s41598-019-56241-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Compared with traditional monolayer cell culture, the three-dimensional tumor spheroid has emerged as an essential in vitro model for cancer research due to the recapitulation of the architecture and physiology of solid human tumors. Herein, by implementing the rapid prototyping of a benchtop 3D printer, we developed a new strategy to generate and analyze tumor spheroids on a commonly used multi-well plate. In this method, the printed artifact can be directly mounted on a 96/384-well plate, enables hanging drop-based spheroid formation, avoiding the tedious fabrication process from micromechanical systems. Besides long-term spheroid culture (20 days), this method supports subsequent analysis of tumor spheroid by seamlessly dripping from the printed array, thereby eliminating the need for spheroids retrieval for downstream characterization. We demonstrated several tumor spheroid-based assays, including tumoroid drug testing, metastasis on or inside extracellular matrix gel, and tumor transendothelial (TEM) assay. Based on quantitative phenotypical and molecular analysis without any precarious retrieval and transfer, we found that the malignant breast cancer (MDA-MB-231) cell aggregate presents a more metastatic morphological phenotype than the non-malignant breast cancer (MCF-7) and colonial cancer (HCT-116) cell spheroid, and shows an up-regulation of epithelial-mesenchymal transition (EMT) relevant genes (fold change > 2). Finally, we validated this tumor malignancy by the TEM assay, which could be easily performed using our approach. This methodology could provide a useful workflow for expediting tumoroid modeled in vitro assay, allowing the “Lab-on-a-Cloud” scenario for routine study.
Collapse
Affiliation(s)
- Liang Zhao
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China. .,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China. .,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China. .,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Jidong Xiu
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China.,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China.,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China.,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yang Liu
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China.,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China.,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China.,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Tianye Zhang
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China.,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China.,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China.,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Wenjie Pan
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China.,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China.,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China.,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Xiaonan Zheng
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China.,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China.,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China.,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Xueji Zhang
- Institute of Precision Medicine and Health, University of Science and Technology Beijing, Beijing, 100083, China. .,Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China. .,Beijing Key Laboratory for Bioengineering and Sensing Technology University of Science and Technology Beijing, Beijing, 100083, China. .,School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
32
|
Sirenko O, Parham F, Dea S, Sodhi N, Biesmans S, Mora-Castilla S, Ryan K, Behl M, Chandy G, Crittenden C, Vargas-Hurlston S, Guicherit O, Gordon R, Zanella F, Carromeu C. Functional and Mechanistic Neurotoxicity Profiling Using Human iPSC-Derived Neural 3D Cultures. Toxicol Sci 2019; 167:58-76. [PMID: 30169818 DOI: 10.1093/toxsci/kfy218] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Neurological disorders affect millions of people worldwide and appear to be on the rise. Whereas the reason for this increase remains unknown, environmental factors are a suspected contributor. Hence, there is an urgent need to develop more complex, biologically relevant, and predictive in vitro assays to screen larger sets of compounds with the potential for neurotoxicity. Here, we employed a human induced pluripotent stem cell (iPSC)-based 3D neural platform composed of mature cortical neurons and astrocytes as a model for this purpose. The iPSC-derived human 3D cortical neuron/astrocyte co-cultures (3D neural cultures) present spontaneous synchronized, readily detectable calcium oscillations. This advanced neural platform was optimized for high-throughput screening in 384-well plates and displays highly consistent, functional performance across different wells and plates. Characterization of oscillation profiles in 3D neural cultures was performed through multi-parametric analysis that included the calcium oscillation rate and peak width, amplitude, and waveform irregularities. Cellular and mitochondrial toxicity were assessed by high-content imaging. For assay characterization, we used a set of neuromodulators with known mechanisms of action. We then explored the neurotoxic profile of a library of 87 compounds that included pharmaceutical drugs, pesticides, flame retardants, and other chemicals. Our results demonstrated that 57% of the tested compounds exhibited effects in the assay. The compounds were then ranked according to their effective concentrations based on in vitro activity. Our results show that a human iPSC-derived 3D neural culture assay platform is a promising biologically relevant tool to assess the neurotoxic potential of drugs and environmental toxicants.
Collapse
Affiliation(s)
| | - Frederick Parham
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Steven Dea
- StemoniX, Inc, Maple Grove, Minnesota 55311
| | - Neha Sodhi
- StemoniX, Inc, Maple Grove, Minnesota 55311
| | | | | | - Kristen Ryan
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Mamta Behl
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Celik SBG, Dominici SR, Filby BW, Das AAK, Madden LA, Paunov VN. Fabrication of Human Keratinocyte Cell Clusters for Skin Graft Applications by Templating Water-in-Water Pickering Emulsions. Biomimetics (Basel) 2019; 4:E50. [PMID: 31336810 PMCID: PMC6784416 DOI: 10.3390/biomimetics4030050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/24/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
Most current methods for the preparation of tissue spheroids require complex materials, involve tedious physical steps and are generally not scalable. We report a novel alternative, which is both inexpensive and up-scalable, to produce large quantities of viable human keratinocyte cell clusters (clusteroids). The method is based on a two-phase aqueous system of incompatible polymers forming a stable water-in-water (w/w) emulsion, which enabled us to rapidly fabricate cell clusteroids from HaCaT cells. We used w/w Pickering emulsion from aqueous solutions of the polymers dextran (DEX) and polyethylene oxide (PEO) and a particle stabilizer based on whey protein (WP). The HaCaT cells clearly preferred to distribute into the DEX-rich phase and this property was utilized to encapsulate them in the water-in-water (DEX-in-PEO) emulsion drops then osmotically shrank to compress them into clusters. Prepared formulations of HaCaT keratinocyte clusteroids in alginate hydrogel were grown where the cells percolated to mimic 3D tissue. The HaCaT cell clusteroids grew faster in the alginate film compared to the individual cells formulated in the same matrix. This methodology could potentially be utilised in biomedical applications.
Collapse
Affiliation(s)
- Sevde B G Celik
- Department of Chemistry and Biochemistry, University of Hull, Hull HU6 7RX, UK
| | | | - Benjamin W Filby
- Department of Chemistry and Biochemistry, University of Hull, Hull HU6 7RX, UK
| | - Anupam A K Das
- Department of Chemistry and Biochemistry, University of Hull, Hull HU6 7RX, UK
| | - Leigh A Madden
- Department of Biomedical Science, University of Hull, Hull HU6 7RX, UK
| | - Vesselin N Paunov
- Department of Chemistry and Biochemistry, University of Hull, Hull HU6 7RX, UK.
| |
Collapse
|
34
|
Spontaneously Formed Spheroids from Mouse Compact Bone-Derived Cells Retain Highly Potent Stem Cells with Enhanced Differentiation Capability. Stem Cells Int 2019; 2019:8469012. [PMID: 31191686 PMCID: PMC6525826 DOI: 10.1155/2019/8469012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 02/07/2023] Open
Abstract
The results from our recent study showed the presence of two distinct spheroid-forming mechanisms, i.e., spontaneous and mechanical. In this study, we focused on the spontaneously formed spheroids, and the character of spontaneously formed spheroids from mouse compact bone-derived cells (CBDCs) was explored. Cells from (C57BL/6J) mouse leg bones were isolated, and compact bone-derived cells were cultured after enzymatic digestion. Spontaneous spheroid formation was achieved on a culture plate with specific water contact angle as reported. The expression levels of embryonic stem cell markers were analyzed using immunofluorescence and quantitative reverse transcription polymerase chain reaction. Then, the cells from spheroids were induced into osteogenic and neurogenic lineages. The spontaneously formed spheroids from CBDCs were positive for ES cell markers such as SSEA1, Sox2, Oct4, and Nanog. Additionally, the expressions of fucosyltransferase 4/FUT4 (SSEA1), Sox2, and Nanog were significantly higher than those in monolayer cultured cells. The gene expression of mesenchymal stem cell markers was almost identical in both spheroids and monolayer-cultured cells, but the expression of Sca-1 was higher in spheroids. Spheroid-derived cells showed significantly higher osteogenic and neurogenic marker expression than monolayer-cultured cells after induction. Spontaneously formed spheroids expressed stem cell markers and showed enhanced osteogenic and neurogenic differentiation capabilities than cells from the conventional monolayer culture, which supports the superior stemness.
Collapse
|
35
|
Lee IC. Cancer-on-a-chip for Drug Screening. Curr Pharm Des 2019; 24:5407-5418. [DOI: 10.2174/1381612825666190206235233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 12/24/2022]
Abstract
:
The oncology pharmaceutical research spent a shocking amount of money on target validation and
drug optimization in preclinical models because many oncology drugs fail during clinical trial phase III. One of
the most important reasons for oncology drug failures in clinical trials may due to the poor predictive tool of
existing preclinical models. Therefore, in cancer research and personalized medicine field, it is critical to improve
the effectiveness of preclinical predictions of the drug response of patients to therapies and to reduce costly failures
in clinical trials. Three dimensional (3D) tumor models combine micro-manufacturing technologies mimic
critical physiologic parameters present in vivo, including complex multicellular architecture with multicellular
arrangement and extracellular matrix deposition, packed 3D structures with cell–cell interactions, such as tight
junctions, barriers to mass transport of drugs, nutrients and other factors, which are similar to in vivo tumor tissues.
These systems provide a solution to mimic the physiological environment for improving predictive accuracy
in oncology drug discovery.
:
his review gives an overview of the innovations, development and limitations of different types of tumor-like
construction techniques such as self-assemble spheroid formation, spheroids formation by micro-manufacturing
technologies, micro-dissected tumor tissues and tumor organoid. Combination of 3D tumor-like construction and
microfluidic techniques to achieve tumor on a chip for in vitro tumor environment modeling and drug screening
were all included. Eventually, developmental directions and technical challenges in the research field are also
discussed. We believe tumor on chip models have provided better sufficient clinical predictive power and will
bridge the gap between proof-of-concept studies and a wider implementation within the oncology drug development
for pathophysiological applications.
Collapse
Affiliation(s)
- I-Chi Lee
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
36
|
Gijbels E, Vilas-Boas V, Deferm N, Devisscher L, Jaeschke H, Annaert P, Vinken M. Mechanisms and in vitro models of drug-induced cholestasis. Arch Toxicol 2019; 93:1169-1186. [PMID: 30972450 DOI: 10.1007/s00204-019-02437-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Cholestasis underlies one of the major manifestations of drug-induced liver injury. Drug-induced cholestatic liver toxicity is a complex process, as it can be triggered by a variety of factors that induce 2 types of biological responses, namely a deteriorative response, caused by bile acid accumulation, and an adaptive response, aimed at removing the accumulated bile acids. Several key events in both types of responses have been characterized in the past few years. In parallel, many efforts have focused on the development and further optimization of experimental cell culture models to predict the occurrence of drug-induced cholestatic liver toxicity in vivo. In this paper, a state-of-the-art overview of mechanisms and in vitro models of drug-induced cholestatic liver injury is provided.
Collapse
Affiliation(s)
- Eva Gijbels
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Vânia Vilas-Boas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Neel Deferm
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, O&N2, Herestraat 49, Bus 921, 3000, Leuven, Belgium
| | - Lindsey Devisscher
- Basic and Applied Medical Sciences, Gut-Liver Immunopharmacology Unit, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, 9000, Ghent, Belgium
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Boulevard, MS 1018, Kansas City, KS, 66160, USA
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, O&N2, Herestraat 49, Bus 921, 3000, Leuven, Belgium
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| |
Collapse
|
37
|
Kim EM, Lee YB, Byun H, Chang HK, Park J, Shin H. Fabrication of Spheroids with Uniform Size by Self-Assembly of a Micro-Scaled Cell Sheet (μCS): The Effect of Cell Contraction on Spheroid Formation. ACS APPLIED MATERIALS & INTERFACES 2019; 11:2802-2813. [PMID: 30586277 DOI: 10.1021/acsami.8b18048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cell spheroid culture can be an effective approach for providing an engineered microenvironment similar to an in vivo environment. Our group had recently developed a method for harvesting uniformly sized multicellular spheroids via self-assembly of micro-scaled cell sheets (μCSs) induced by the expansion of temperature-sensitive hydrogels. However, the μCS assembly process was not fully understood. In this study, we investigated the effects of cell number, pattern shape, and contractile force of cells on spheroid formation from micropatterned (width of square pattern from 100-300 μm) hydrogels. We used human dermal fibroblasts (HDFBs) as a model cell type and cultured them for 24 and 72 h. The self-assembly of μCSs cultured on square micropatterns for 72 h rapidly occurred within 4 min after reducing the temperature from 37 to 4 °C. In addition, the size distribution of spheroids was narrower with μCSs from a 72 h culture. Treatment with a ROCK1 inhibitor disrupted cytoskeletal actin fibers and the corresponding μCSs were not detached from the hydrogel. The assembly of the μCS was also affected by the micropattern shape, and the spheroid harvest efficiency was decreased to 60% when using a circular micropattern, which was explained by the stress direction on the circular versus square micropattern upon hydrogel expansion. Therefore, we confirmed that the factors controlling cell-cell interactions are important for spheroid formation using micropatterned hydrogel systems. Finally, the μCSs with dual layers of HDFBs labeled with DiD and DiO dyes resulted in the formation of spheroids with discretely localized colors within the core and shell, respectively, which suggests an outside-in assembly of detached μCSs. In consideration of these complex environmental factors, our system could be utilized in various applications as a three-dimensional culture system to fabricate cell spheroids.
Collapse
Affiliation(s)
| | | | | | - Hyung-Kwan Chang
- Department of Mechanical Engineering , Sogang University , 35 Baekbeom-ro , Mapo-gu, Seoul 04107 , Republic of Korea
| | - Jungyul Park
- Department of Mechanical Engineering , Sogang University , 35 Baekbeom-ro , Mapo-gu, Seoul 04107 , Republic of Korea
| | | |
Collapse
|
38
|
Laternser S, Keller H, Leupin O, Rausch M, Graf-Hausner U, Rimann M. A Novel Microplate 3D Bioprinting Platform for the Engineering of Muscle and Tendon Tissues. SLAS Technol 2018; 23:599-613. [PMID: 29895208 PMCID: PMC6249648 DOI: 10.1177/2472630318776594] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/09/2018] [Accepted: 04/16/2018] [Indexed: 12/16/2022]
Abstract
Two-dimensional (2D) cell cultures do not reflect the in vivo situation, and thus it is important to develop predictive three-dimensional (3D) in vitro models with enhanced reliability and robustness for drug screening applications. Treatments against muscle-related diseases are becoming more prominent due to the growth of the aging population worldwide. In this study, we describe a novel drug screening platform with automated production of 3D musculoskeletal-tendon-like tissues. With 3D bioprinting, alternating layers of photo-polymerized gelatin-methacryloyl-based bioink and cell suspension tissue models were produced in a dumbbell shape onto novel postholder cell culture inserts in 24-well plates. Monocultures of human primary skeletal muscle cells and rat tenocytes were printed around and between the posts. The cells showed high viability in culture and good tissue differentiation, based on marker gene and protein expressions. Different printing patterns of bioink and cells were explored and calcium signaling with Fluo4-loaded cells while electrically stimulated was shown. Finally, controlled co-printing of tenocytes and myoblasts around and between the posts, respectively, was demonstrated followed by co-culture and co-differentiation. This screening platform combining 3D bioprinting with a novel microplate represents a promising tool to address musculoskeletal diseases.
Collapse
Affiliation(s)
- Sandra Laternser
- Competence Center TEDD, Institute of
Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences,
Waedenswil, Switzerland
- Center for Cell Biology & Tissue
Engineering, Institute of Chemistry and Biotechnology (ICBT), Zurich University of
Applied Sciences, Waedenswil, Switzerland
| | - Hansjoerg Keller
- Musculoskeletal Diseases, Novartis
Institutes for BioMedical Research, Basel, Switzerland
| | - Olivier Leupin
- Musculoskeletal Diseases, Novartis
Institutes for BioMedical Research, Basel, Switzerland
| | - Martin Rausch
- Biotherapeutic and Analytical
Technologies, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ursula Graf-Hausner
- Competence Center TEDD, Institute of
Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences,
Waedenswil, Switzerland
- Center for Cell Biology & Tissue
Engineering, Institute of Chemistry and Biotechnology (ICBT), Zurich University of
Applied Sciences, Waedenswil, Switzerland
| | - Markus Rimann
- Competence Center TEDD, Institute of
Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences,
Waedenswil, Switzerland
- Center for Cell Biology & Tissue
Engineering, Institute of Chemistry and Biotechnology (ICBT), Zurich University of
Applied Sciences, Waedenswil, Switzerland
| |
Collapse
|
39
|
Cui X, Hartanto Y, Wu C, Bi J, Dai S, Zhang H. Tuning microenvironment for multicellular spheroid formation in thermo‐responsive anionic microgel scaffolds. J Biomed Mater Res A 2018; 106:2899-2909. [DOI: 10.1002/jbm.a.36479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Xiaolin Cui
- School of Chemical Engineering the University of Adelaide Adelaide Australia
| | - Yusak Hartanto
- School of Chemical Engineering the University of Adelaide Adelaide Australia
| | - Chengtie Wu
- Biomaterials and Tissue Engineering Research Centre, Shanghai Institute of Ceramics, Chinese Academy of Sciences Shanghai China
| | - Jingxiu Bi
- School of Chemical Engineering the University of Adelaide Adelaide Australia
| | - Sheng Dai
- School of Chemical Engineering and Advanced Materials Newcastle University Newcastle‐upon‐Tyne United Kingdom
| | - Hu Zhang
- School of Chemical Engineering the University of Adelaide Adelaide Australia
| |
Collapse
|
40
|
Impact of Three-Dimentional Culture Systems on Hepatic Differentiation of Puripotent Stem Cells and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018. [PMID: 30357683 DOI: 10.1007/978-981-13-0947-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Generation of functional hepatocytes from human pluripotent stem cells (hPSCs) is a vital tool to produce large amounts of human hepatocytes, which hold a great promise for biomedical and regenerative medicine applications. Despite a tremendous progress in developing the differentiation protocols recapitulating the developmental signalling and stages, these resulting hepatocytes from hPSCs yet achieve maturation and functionality comparable to those primary hepatocytes. The absence of 3D milieu in the culture and differentiation of these hepatocytes may account for this, at least partly, thus developing an optimal 3D culture could be a step forward to achieve this aim. Hence, review focuses on current development of 3D culture systems for hepatic differentiation and maturation and the future perspectives of its application.
Collapse
|
41
|
Wolf CJ, Belair DG, Becker CM, Das KP, Schmid JE, Abbott BD. Development of an organotypic stem cell model for the study of human embryonic palatal fusion. Birth Defects Res 2018; 110:1322-1334. [PMID: 30347137 DOI: 10.1002/bdr2.1394] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Cleft palate (CP) is a common birth defect, occurring in an estimated 1 in 1,000 births worldwide. The secondary palate is formed by paired palatal shelves, consisting of a mesenchymal core with an outer layer of epithelial cells that grow toward each other, attach, and fuse. One of the mechanisms that can cause CP is failure of fusion, that is, failure to remove the epithelial seam between the palatal shelves to allow the mesenchyme confluence. Epidermal growth factor (EGF) plays an important role in palate growth and differentiation, while it may impede fusion. METHODS We developed a 3D organotypic model using human mesenchymal and epithelial stem cells to mimic human embryonic palatal shelves, and tested the effects of human EGF (hEGF) on proliferation and fusion. Spheroids were generated from human umbilical-derived mesenchymal stem cells (hMSCs) directed down an osteogenic lineage. Heterotypic spheroids, or organoids, were constructed by coating hMSC spheroids with extracellular matrix solution followed by a layer of human progenitor epithelial keratinocytes (hPEKs). Organoids were incubated in co-culture medium with or without hEGF and assessed for cell proliferation and time to fusion. RESULTS Osteogenic differentiation in hMSC spheroids was highest by Day 13. hEGF delayed fusion of organoids after 12 and 18 hr of contact. hEGF increased proliferation in organoids at 4 ng/ml, and proliferation was detected in hPEKs alone. CONCLUSION Our results show that this model of human palatal fusion appropriately mimics the morphology of the developing human palate and responds to hEGF as expected.
Collapse
Affiliation(s)
- Cynthia J Wolf
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - David G Belair
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - Carrie M Becker
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee
| | - Kaberi P Das
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - Judith E Schmid
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| | - Barbara D Abbott
- Toxicity Assessment Division, National Health and Environmental Effects Research Laboratories, Office of Research and Development, US EPA Research Triangle Park, North Carolina
| |
Collapse
|
42
|
Zhai Q, Dong Z, Wang W, Li B, Jin Y. Dental stem cell and dental tissue regeneration. Front Med 2018; 13:152-159. [PMID: 29971640 DOI: 10.1007/s11684-018-0628-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/14/2017] [Indexed: 12/22/2022]
Abstract
The teeth are highly differentiated chewing organs formed by the development of tooth germ tissue located in the jaw and consist of the enamel, dentin, cementum, pulp, and periodontal tissue. Moreover, the teeth have a complicated regulatory mechanism, special histologic origin, diverse structure, and important function in mastication, articulation, and aesthetics. These characteristics, to a certain extent, greatly complicate the research in tooth regeneration. Recently, new ideas for tooth and tissue regeneration have begun to appear with rapid developments in the theories and technologies in tissue engineering. Numerous types of stem cells have been isolated from dental tissue, such as dental pulp stem cells (DPSCs), stem cells isolated from human pulp of exfoliated deciduous teeth (SHED), periodontal ligament stem cells (PDLSCs), stem cells from apical papilla (SCAPs), and dental follicle cells (DFCs). All these cells can regenerate the tissue of tooth. This review outlines the cell types and strategies of stem cell therapy applied in tooth regeneration, in order to provide theoretical basis for clinical treatments.
Collapse
Affiliation(s)
- Qiming Zhai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhiwei Dong
- Department of Oral and Maxillofacial Surgery, General Hospital of Shenyang Military Area Command, Shenyang, 110840, China
| | - Wei Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Bei Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, 710032, China. .,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, 710032, China. .,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China.
| |
Collapse
|
43
|
Challenges in Bio-fabrication of Organoid Cultures. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:53-71. [DOI: 10.1007/5584_2018_216] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
44
|
Aeby EA, Misun PM, Hierlemann A, Frey O. Microfluidic Hydrogel Hanging-Drop Network for Long-Term Culturing of 3D Microtissues and Simultaneous High-Resolution Imaging. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Elise A. Aeby
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| | - Patrick M. Misun
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| | - Olivier Frey
- Bio Engineering Laboratory; Department of Biosystems Science and Engineering; ETH Zürich; Mattenstrasse 26 4058 Basel Switzerland
| |
Collapse
|
45
|
Fakoya AOJ, Otohinoyi DA, Yusuf J. Current Trends in Biomaterial Utilization for Cardiopulmonary System Regeneration. Stem Cells Int 2018; 2018:3123961. [PMID: 29853910 PMCID: PMC5949153 DOI: 10.1155/2018/3123961] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/15/2017] [Accepted: 03/01/2018] [Indexed: 12/28/2022] Open
Abstract
The cardiopulmonary system is made up of the heart and the lungs, with the core function of one complementing the other. The unimpeded and optimal cycling of blood between these two systems is pivotal to the overall function of the entire human body. Although the function of the cardiopulmonary system appears uncomplicated, the tissues that make up this system are undoubtedly complex. Hence, damage to this system is undesirable as its capacity to self-regenerate is quite limited. The surge in the incidence and prevalence of cardiopulmonary diseases has reached a critical state for a top-notch response as it currently tops the mortality table. Several therapies currently being utilized can only sustain chronically ailing patients for a short period while they are awaiting a possible transplant, which is also not devoid of complications. Regenerative therapeutic techniques now appear to be a potential approach to solve this conundrum posed by these poorly self-regenerating tissues. Stem cell therapy alone appears not to be sufficient to provide the desired tissue regeneration and hence the drive for biomaterials that can support its transplantation and translation, providing not only physical support to seeded cells but also chemical and physiological cues to the cells to facilitate tissue regeneration. The cardiac and pulmonary systems, although literarily seen as just being functionally and spatially cooperative, as shown by their diverse and dissimilar adult cellular and tissue composition has been proven to share some common embryological codevelopment. However, necessitating their consideration for separate review is the immense adult architectural difference in these systems. This review also looks at details on new biological and synthetic biomaterials, tissue engineering, nanotechnology, and organ decellularization for cardiopulmonary regenerative therapies.
Collapse
Affiliation(s)
| | | | - Joshua Yusuf
- All Saints University School of Medicine, Roseau, Dominica
- All Saints University School of Medicine, Kingstown, Saint Vincent and the Grenadines
| |
Collapse
|
46
|
Maji S, Agarwal T, Das J, Maiti TK. Development of gelatin/carboxymethyl chitosan/nano-hydroxyapatite composite 3D macroporous scaffold for bone tissue engineering applications. Carbohydr Polym 2018; 189:115-125. [PMID: 29580388 DOI: 10.1016/j.carbpol.2018.01.104] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 12/26/2022]
Abstract
The present study delineates a relatively simpler approach for fabrication of a macroporous three-dimensional scaffold for bone tissue engineering. The novelty of the work is to obtain a scaffold with macroporosity (interconnected networks) through a combined approach of high stirring induced foaming of the gelatin/carboxymethyl chitosan (CMC)/nano-hydroxyapatite (nHAp) matrix followed by freeze drying. The fabricated macroporous (SGC) scaffold had a greater pore size, higher porosity, higher water retention capacity, slow and sustained enzymatic degradation rate along with higher compressive strength compared to that of non-macroporous (NGC, prepared by conventional freeze drying methodology) scaffold. The biological studies revealed the increased percentage of viability, proliferation, and differentiation as well as higher mineralization of differentiated human Wharton's jelly MSC microtissue (wjhMSC-MT) on SGC as compared to NGC scaffold. RT-PCR also showed enhanced expression level of collagen type I, osteocalcin and Runx2 when seeded on SGC. μCT and histological analysis further revealed a penetration of cellular spheroid to a greater depth in SGC scaffold than NGC scaffold. Furthermore, the effect of cryopreservation on microtissue survival on the three-dimensional construct revealed significant higher viability upon revival in macroporous SGC scaffolds. These results together suggest that high stirring based macroporous scaffolds could have a potential application in bone tissue engineering.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| | - Joyjyoti Das
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur-721302, West Bengal, India.
| |
Collapse
|
47
|
Kyffin JA, Sharma P, Leedale J, Colley HE, Murdoch C, Mistry P, Webb SD. Impact of cell types and culture methods on the functionality of in vitro liver systems - A review of cell systems for hepatotoxicity assessment. Toxicol In Vitro 2018; 48:262-275. [PMID: 29408671 DOI: 10.1016/j.tiv.2018.01.023] [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] [Received: 09/26/2017] [Revised: 01/26/2018] [Accepted: 01/27/2018] [Indexed: 12/21/2022]
Abstract
Xenobiotic safety assessment is an area that impacts a multitude of different industry sectors such as medicinal drugs, agrochemicals, industrial chemicals, cosmetics and environmental contaminants. As such there are a number of well-developed in vitro, in vivo and in silico approaches to evaluate their properties and potential impact on the environment and to humans. Additionally, there is the continual investment in multidisciplinary scientists to explore non-animal surrogate technologies to predict specific toxicological outcomes and to improve our understanding of the biological processes regarding the toxic potential of xenobiotics. Here we provide a concise, critical evaluation of a number of in vitro systems utilised to assess the hepatotoxic potential of xenobiotics.
Collapse
Affiliation(s)
- Jonathan A Kyffin
- Department of Applied Mathematics, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Parveen Sharma
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Sherrington Building, Ashton Street, University of Liverpool, L69 3GE, United Kingdom.
| | - Joseph Leedale
- EPSRC Liverpool Centre for Mathematics in Healthcare, Department of Mathematical Sciences, Peach Street, University of Liverpool, L69 7ZL, United Kingdom
| | - Helen E Colley
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Craig Murdoch
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, Sheffield S10 2TA, United Kingdom
| | - Pratibha Mistry
- Syngenta Ltd., Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, United Kingdom
| | - Steven D Webb
- Department of Applied Mathematics, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, United Kingdom
| |
Collapse
|
48
|
Alexander F, Eggert S, Wiest J. A novel lab-on-a-chip platform for spheroid metabolism monitoring. Cytotechnology 2018; 70:375-386. [PMID: 29032507 PMCID: PMC5809666 DOI: 10.1007/s10616-017-0152-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/04/2017] [Indexed: 12/24/2022] Open
Abstract
Sensor-based cellular microphysiometry is a technique that allows non-invasive, label-free, real-time monitoring of living cells that can greatly improve the predictability of toxicology testing by removing the influence of biochemical labels. In this work, the Intelligent Mobile Lab for In Vitro Diagnostics (IMOLA-IVD) was utilized to perform cellular microphysiometry on 3D multicellular spheroids. Using a commercial 3D printer, 3 × 3 microwell arrays were fabricated to maintain nine previously cultured HepG2 spheroids on a single BioChip. Integrated layers above and under the spheroids allowed fluidic contact between spheroids in microwells and BioChip sensors while preventing wash out from medium perfusion. Spheroid culturing protocols were optimized to grow spheroids to a diameter of around 620 μm prior to transfer onto BioChips. An ON/OFF pump cycling protocol was developed to optimize spheroid culture within the designed microwells, intermittently perfuse spheroids with fresh culture medium, and measure the extracellular acidification rate (EAR) and oxygen uptake rate (OUR) with the BioChips of the IMOLA-IVD platform. In a proof-of-concept experiment, spheroids were perfused for 36 h with cell culture medium before being exposed to medium with 1% sodium dodecyl sulphate (SDS) to lyse cells as a positive control. These microphysiometry studies revealed a repeatable pattern of extracellular acidification throughout the experiment, indicating the ability to monitor real-time metabolic activity of spheroids embedded in the newly designed tissue encapsulation. After perfusion for 36 h with medium, SDS exposure resulted in an instant decrease in EAR and OUR signals from 37 mV/h (± 5) to 8 mV/h (± 8) and from 308 mV/h (± 21) to -2 mV/h (± 13), respectively. The presented spheroid monitoring system holds great potential as a method to automate screening and analysis of pharmaceutical agents using 3D multicellular spheroid models.
Collapse
Affiliation(s)
| | - Sebastian Eggert
- cellasys GmbH - R&D, Ohmstraße 8, 80802, Munich, Germany
- Technical University of Munich, Arcisstraße 21, 80333, Munich, Germany
| | - Joachim Wiest
- cellasys GmbH - R&D, Ohmstraße 8, 80802, Munich, Germany.
| |
Collapse
|
49
|
Wang J, Miao Y, Huang Y, Lin B, Liu X, Xiao S, Du L, Hu Z, Xing M. Bottom-up Nanoencapsulation from Single Cells to Tunable and Scalable Cellular Spheroids for Hair Follicle Regeneration. Adv Healthc Mater 2018; 7. [PMID: 29227036 DOI: 10.1002/adhm.201700447] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/05/2017] [Indexed: 01/31/2023]
Abstract
Cell surface engineering technology advances cell therapeutics and tissue engineering by accurate micro/nanoscale control in cell-biomaterial ensembles and cell spheroids formation. By tailoring cell surface, microgels can encapsulate cells for versatile uses. However, microgels are coated in a thick layer to house multiple cells together but not a single cell based. Besides, excessive deposition on cell surface is detrimental to cellular functions. Herein, layer-by-layer (LbL) self-assembly to encapsulate single cell using nanogel is reported, owing to its security and tunable thickness at nanoscale, and further forms cell spheroids by physical cross-linking on nanogel-coated cells for delivery. A hair follicle (HF) regeneration model where the dermal papilla cells (DPCs) are given a 3D installation to maintain its ability of HF induction during in vitro culture is studied. Dermal papilla (DP) spheroids are optimized and that LbL-DPCs aggregation is akin to primary DP is demonstrated. The markers ALP, Versican, and NCAM are examined to investigate that high-passaged (P8) DP spheroids can restore the hair induction potential, which are lost in 2D culture. New HFs are regenerated successfully by implantation of DP spheroids in vivo.
Collapse
Affiliation(s)
- Jin Wang
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
- Department of Mechanical Engineering; University of Manitoba; 75A Chancellors Circle Winnipeg Manitoba R3T 2N2 Canada
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
| | - Yong Huang
- Chongqing Academy of Animal Sciences; Chongqing 402460 China
| | - Bojie Lin
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
- Department of Mechanical Engineering; University of Manitoba; 75A Chancellors Circle Winnipeg Manitoba R3T 2N2 Canada
| | - Xiaomin Liu
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
| | - Shune Xiao
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
| | - Lijuan Du
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery; Nanfang Hospital of Southern Medical University; Guangzhou Guangdong Province 510515 China
| | - Malcolm Xing
- Department of Mechanical Engineering; University of Manitoba; 75A Chancellors Circle Winnipeg Manitoba R3T 2N2 Canada
- Children's Hospital Research Institute of Manitoba; 715 McDermot Ave Winnipeg Manitoba R3E3P4 Canada
| |
Collapse
|
50
|
Lei J, Murphy WL, Temenoff JS. Combination of Heparin Binding Peptide and Heparin Cell Surface Coatings for Mesenchymal Stem Cell Spheroid Assembly. Bioconjug Chem 2018; 29:878-884. [PMID: 29341600 DOI: 10.1021/acs.bioconjchem.7b00757] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Microtissues containing multiple cell types have been used in both in vitro models and in vivo tissue repair applications. However, to improve throughput, there is a need to develop a platform that supports self-assembly of a large number of 3D microtissues containing multiple cell types in a dynamic suspension system. Thus, the objective of this study was to exploit the binding interaction between the negatively charged glycosaminoglycan, heparin, and a known heparin binding peptide to establish a method that promotes assembly of mesenchymal stem cell (MSC) spheroids into larger aggregates. We characterized heparin binding peptide (HEPpep) and heparin coatings on cell surfaces and determined the specificity of these coatings in promoting assembly of MSC spheroids in dynamic culture. Overall, combining spheroids with both coatings promoted up to 70 ± 11% of spheroids to assemble into multiaggregate structures, as compared to only 10 ± 4% assembly when cells having the heparin coating were cultured with cells coated with a scrambled peptide. These results suggest that this self-assembly method represents an exciting approach that may be applicable for a wide range of applications in which cell aggregation is desired.
Collapse
Affiliation(s)
| | - William L Murphy
- Department of Biomedical Engineering , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States.,Department of Orthopedics and Rehabilitation , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Johnna S Temenoff
- Coulter Department of Biomedical Engineering , Georgia Tech/Emory University , Atlanta , Georgia 30332 , United States
| |
Collapse
|