1
|
Schmitz MGJ, Aarts JGM, Burroughs L, Sudarsanam P, Kuijpers TJM, Riool M, de Boer L, Xue X, Bosnacki D, Zaat SAJ, de Boer J, Alexander MR, Dankers PYW. Merging Modular Molecular Design with High Throughput Screening of Cell Adhesion on Antimicrobial Supramolecular Biomaterials. Macromol Rapid Commun 2025; 46:e2300638. [PMID: 38530968 PMCID: PMC12004893 DOI: 10.1002/marc.202300638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/05/2024] [Indexed: 03/28/2024]
Abstract
A polymer microarray based on the supramolecular ureido-pyrimidinone (UPy) moiety is fabricated to screen antimicrobial materials for their ability to support cell adhesion. UPy-functionalized additives, either cell-adhesive, antimicrobial or control peptides, are used, and investigated in different combinations at different concentrations, resulting in a library of 194 spots. These are characterized on composition and morphology to evaluate the microarray fabrication. Normal human dermal fibroblasts are cultured on the microarrays and cell adhesion to the spots is systematically analyzed. Results demonstrate enhanced cell adhesion on spots with combinations including the antimicrobial peptides. This study clearly proves the power of the high throughput approach in combination with supramolecular molecules, to screen additive libraries for desired biological response.
Collapse
Affiliation(s)
- Moniek G. J. Schmitz
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| | - Jasper G. M. Aarts
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| | - Laurence Burroughs
- School of PharmacyUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
| | - Phanikrishna Sudarsanam
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| | - Tim J. M. Kuijpers
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| | - Martijn Riool
- Department of Medical Microbiology and Infection PreventionAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdam1105 AZThe Netherlands
- Present address:
Laboratory of Experimental Trauma Surgery, Department of Trauma SurgeryUniversity Hospital RegensburgAm Biopark 993053RegensburgGermany
| | - Leonie de Boer
- Department of Medical Microbiology and Infection PreventionAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdam1105 AZThe Netherlands
| | - Xuan Xue
- School of PharmacyUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
| | - Dragan Bosnacki
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| | - Sebastian A. J. Zaat
- Department of Medical Microbiology and Infection PreventionAmsterdam institute for Infection and ImmunityAmsterdam University Medical CentersUniversity of AmsterdamAmsterdam1105 AZThe Netherlands
| | - Jan de Boer
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| | - Morgan R. Alexander
- School of PharmacyUniversity of NottinghamUniversity ParkNottinghamNG7 2RDUK
| | - Patricia Y. W. Dankers
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
| |
Collapse
|
2
|
Zhang W, Rao Y, Wong SH, Wu Y, Zhang Y, Yang R, Tsui SK, Ker DFE, Mao C, Frith JE, Cao Q, Tuan RS, Wang DM. Transcriptome-Optimized Hydrogel Design of a Stem Cell Niche for Enhanced Tendon Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2313722. [PMID: 39417770 PMCID: PMC11733723 DOI: 10.1002/adma.202313722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Bioactive hydrogels have emerged as promising artificial niches for enhancing stem cell-mediated tendon repair. However, a substantial knowledge gap remains regarding the optimal combination of niche features for targeted cellular responses, which often leads to lengthy development cycles and uncontrolled healing outcomes. To address this critical gap, an innovative, data-driven materiomics strategy is developed. This approach is based on in-house RNA-seq data that integrates bioinformatics and mathematical modeling, which is a significant departure from traditional trial-and-error methods. It aims to provide both mechanistic insights and quantitative assessments and predictions of the tenogenic effects of adipose-derived stem cells induced by systematically modulated features of a tendon-mimetic hydrogel (TenoGel). The knowledge generated has enabled a rational approach for TenoGel design, addressing key considerations, such as tendon extracellular matrix concentration, uniaxial tensile loading, and in vitro pre-conditioning duration. Remarkably, our optimized TenoGel demonstrated robust tenogenesis in vitro and facilitated tendon regeneration while preventing undesired ectopic ossification in a rat tendon injury model. These findings shed light on the importance of tailoring hydrogel features for efficient tendon repair. They also highlight the tremendous potential of the innovative materiomics strategy as a powerful predictive and assessment tool in biomaterial development for regenerative medicine.
Collapse
Affiliation(s)
- Wanqi Zhang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Ying Rao
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Shing Hei Wong
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Yeung Wu
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Yuanhao Zhang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Rui Yang
- Department of Sports MedicineOrthopedicsSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Stephen Kwok‐Wing Tsui
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Dai Fei Elmer Ker
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongHong Kong SARChina
| | - Jessica E. Frith
- Materials Science and EngineeringMonash UniversityClayton3800VICAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClayton3800VICAustralia
- Australian Research Council Training Centre for Cell and Tissue Engineering TechnologiesMonash UniversityClayton3800VICAustralia
| | - Qin Cao
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Hong Kong Bioinformatics CentreThe Chinese University of Hong KongHong Kong SARChina
| | - Rocky S. Tuan
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Dan Michelle Wang
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- Institute for Tissue Engineering and Regenerative MedicineThe Chinese University of Hong KongHong Kong SARChina
- Center for Neuromusculoskeletal Restorative MedicineHong Kong Science ParkHong Kong SARChina
- Department of Orthopaedics and TraumatologyFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| |
Collapse
|
3
|
Li Z, Song P, Li G, Han Y, Ren X, Bai L, Su J. AI energized hydrogel design, optimization and application in biomedicine. Mater Today Bio 2024; 25:101014. [PMID: 38464497 PMCID: PMC10924066 DOI: 10.1016/j.mtbio.2024.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
Traditional hydrogel design and optimization methods usually rely on repeated experiments, which is time-consuming and expensive, resulting in a slow-moving of advanced hydrogel development. With the rapid development of artificial intelligence (AI) technology and increasing material data, AI-energized design and optimization of hydrogels for biomedical applications has emerged as a revolutionary breakthrough in materials science. This review begins by outlining the history of AI and the potential advantages of using AI in the design and optimization of hydrogels, such as prediction and optimization of properties, multi-attribute optimization, high-throughput screening, automated material discovery, optimizing experimental design, and etc. Then, we focus on the various applications of hydrogels supported by AI technology in biomedicine, including drug delivery, bio-inks for advanced manufacturing, tissue repair, and biosensors, so as to provide a clear and comprehensive understanding of researchers in this field. Finally, we discuss the future directions and prospects, and provide a new perspective for the research and development of novel hydrogel materials for biomedical applications.
Collapse
Affiliation(s)
- Zuhao Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Yafei Han
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiaoxiang Ren
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
4
|
Liu S, Cao C, Wang Y, Hu L, Liu Q. Novel Therapies for ANCA-associated Vasculitis: Apilimod Ameliorated Endothelial Cells Injury through TLR4/NF-κB Pathway and NLRP3 Inflammasome. Curr Pharm Des 2024; 30:2325-2344. [PMID: 38910483 DOI: 10.2174/0113816128312530240607051608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Antineutrophil cytoplasmic antibody-associated vasculitis (AAV) is a rapidly progressive form of glomerulonephritis for which effective therapeutic drugs are currently lacking, and its underlying mechanism remains unclear. AIMS This study aimed to investigate new treatment options for AAV through a combination of bioinformatics analysis and cell molecular experiments. METHODS The research utilized integrated bioinformatics analysis to identify genes with differential expression, conduct enrichment analysis, and pinpoint hub genes associated with AAV. Potential therapeutic compounds for AAV were identified using Connectivity Map and molecular docking techniques. In vitro experiments were then carried out to examine the impact and mechanism of apilimod on endothelial cell injury induced by MPO-ANCA-positive IgG. RESULTS The findings revealed a set of 374 common genes from differentially expressed genes and key modules of WGCNA, which were notably enriched in immune and inflammatory response processes. A proteinprotein interaction network was established, leading to the identification of 10 hub genes, including TYROBP, PTPRC, ITGAM, KIF20A, CD86, CCL20, GAD1, LILRB2, CD8A, and COL5A2. Analysis from Connectivity Map and molecular docking suggested that apilimod could serve as a potential therapeutic cytokine inhibitor for ANCA-GN based on the hub genes. In vitro experiments demonstrated that apilimod could mitigate tight junction disruption, endothelial cell permeability, LDH release, and endothelial activation induced by MPO-ANCA-positive IgG. Additionally, apilimod treatment led to a significant reduction in the expression of proteins involved in the TLR4/NF-κB and NLRP3 inflammasome-mediated pyroptosis pathways. CONCLUSION This study sheds light on the potential pathogenesis of AAV and highlights the protective role of apilimod in mitigating MPO-ANCA-IgG-induced vascular endothelial cell injury by modulating the TLR4/ NF-kB and NLRP3 inflammasome-mediated pyroptosis pathway. These findings suggest that apilimod may hold promise as a treatment for AAV and warrant further investigation.
Collapse
Affiliation(s)
- Siyang Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlin Cao
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of the Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiru Wang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Hu
- Department of Health Management Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Owh C, Ow V, Lin Q, Wong JHM, Ho D, Loh XJ, Xue K. Bottom-up design of hydrogels for programmable drug release. BIOMATERIALS ADVANCES 2022; 141:213100. [PMID: 36096077 DOI: 10.1016/j.bioadv.2022.213100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/22/2022] [Accepted: 08/27/2022] [Indexed: 06/15/2023]
Abstract
Hydrogels are a promising drug delivery system for biomedical applications due to their biocompatibility and similarity to native tissue. Programming the release rate from hydrogels is critical to ensure release of desired dosage over specified durations, particularly with the advent of more complicated medical regimens such as combinatorial drug therapy. While it is known how hydrogel structure affects release, the parameters that can be explicitly controlled to modulate release ab initio could be useful for hydrogel design. In this review, we first survey common physical models of hydrogel release. We then extensively go through the various input parameters that we can exercise direct control over, at the levels of synthesis, formulation, fabrication and environment. We also illustrate some examples where hydrogels can be programmed with the input parameters for temporally and spatially defined release. Finally, we discuss the exciting potential and challenges for programming release, and potential implications with the advent of machine learning.
Collapse
Affiliation(s)
- Cally Owh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore (NUS), 21 Lower Kent Ridge Rd, Singapore 119077, Singapore
| | - Valerie Ow
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Singapore
| | - Qianyu Lin
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore (NUS), 21 Lower Kent Ridge Rd, Singapore 119077, Singapore
| | - Joey Hui Min Wong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Singapore
| | - Dean Ho
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, Singapore 117583, Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Singapore; Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117575, Singapore; School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, #01-30 General Office, Block N4.1, Singapore 639798, Singapore.
| | - Kun Xue
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Singapore.
| |
Collapse
|
6
|
Computational Study of Methods for Determining the Elasticity of Red Blood Cells Using Machine Learning. Symmetry (Basel) 2022. [DOI: 10.3390/sym14081732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RBC (Red Blood Cell) membrane is a highly elastic structure, and proper modelling of this elasticity is essential for biomedical applications that involve computational experiments with blood flow. In this work, we present a new method for estimating one of the key parameters of red blood cell elasticity, which uses a neural network trained on the simulation outputs. We test classic LSTM (Long-Short Term Memory) architecture for the time series regression task, and we also experiment with novel CNN-LSTM (Convolutional Neural Network) architecture. We paid special attention to investigating the impact of the way the three-dimensional training data are reduced to their two-dimensional projections. Such a comparison is possible thanks to working with simulation outputs that are equivalently defined for all dimensions and their combinations. The obtained results can be used as recommendations for an appropriate way to record real experiments for which the reduced dimension of the acquired data is essential.
Collapse
|
7
|
Kumar R. Materiomically Designed Polymeric Vehicles for Nucleic Acids: Quo Vadis? ACS APPLIED BIO MATERIALS 2022; 5:2507-2535. [PMID: 35642794 DOI: 10.1021/acsabm.2c00346] [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: 11/28/2022]
Abstract
Despite rapid advances in molecular biology, particularly in site-specific genome editing technologies, such as CRISPR/Cas9 and base editing, financial and logistical challenges hinder a broad population from accessing and benefiting from gene therapy. To improve the affordability and scalability of gene therapy, we need to deploy chemically defined, economical, and scalable materials, such as synthetic polymers. For polymers to deliver nucleic acids efficaciously to targeted cells, they must optimally combine design attributes, such as architecture, length, composition, spatial distribution of monomers, basicity, hydrophilic-hydrophobic phase balance, or protonation degree. Designing polymeric vectors for specific nucleic acid payloads is a multivariate optimization problem wherein even minuscule deviations from the optimum are poorly tolerated. To explore the multivariate polymer design space rapidly, efficiently, and fruitfully, we must integrate parallelized polymer synthesis, high-throughput biological screening, and statistical modeling. Although materiomics approaches promise to streamline polymeric vector development, several methodological ambiguities must be resolved. For instance, establishing a flexible polymer ontology that accommodates recent synthetic advances, enforcing uniform polymer characterization and data reporting standards, and implementing multiplexed in vitro and in vivo screening studies require considerable planning, coordination, and effort. This contribution will acquaint readers with the challenges associated with materiomics approaches to polymeric gene delivery and offers guidelines for overcoming these challenges. Here, we summarize recent developments in combinatorial polymer synthesis, high-throughput screening of polymeric vectors, omics-based approaches to polymer design, barcoding schemes for pooled in vitro and in vivo screening, and identify materiomics-inspired research directions that will realize the long-unfulfilled clinical potential of polymeric carriers in gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemical & Biological Engineering, Colorado School of Mines, 1613 Illinois St, Golden, Colorado 80401, United States
| |
Collapse
|
8
|
Post JN, Loerakker S, Merks R, Carlier A. Implementing computational modeling in tissue engineering: where disciplines meet. Tissue Eng Part A 2022; 28:542-554. [PMID: 35345902 DOI: 10.1089/ten.tea.2021.0215] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In recent years, the mathematical and computational sciences have developed novel methodologies and insights that can aid in designing advanced bioreactors, microfluidic set-ups or organ-on-chip devices, in optimizing culture conditions, or predicting long-term behavior of engineered tissues in vivo. In this review, we introduce the concept of computational models and how they can be integrated in an interdisciplinary workflow for Tissue Engineering and Regenerative Medicine (TERM). We specifically aim this review of general concepts and examples at experimental scientists with little or no computational modeling experience. We also describe the contribution of computational models in understanding TERM processes and in advancing the TERM field by providing novel insights.
Collapse
Affiliation(s)
- Janine Nicole Post
- University of Twente, 3230, Tissue Regeneration, Enschede, Overijssel, Netherlands;
| | - Sandra Loerakker
- Eindhoven University of Technology, 3169, Department of Biomedical Engineering, Eindhoven, Noord-Brabant, Netherlands.,Eindhoven University of Technology, 3169, Institute for Complex Molecular Systems, Eindhoven, Noord-Brabant, Netherlands;
| | - Roeland Merks
- Leiden University, 4496, Institute for Biology Leiden and Mathematical Institute, Leiden, Zuid-Holland, Netherlands;
| | - Aurélie Carlier
- Maastricht University, 5211, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER Maastricht, Maastricht, Netherlands, 6200 MD;
| |
Collapse
|
9
|
Vermeulen S, Honig F, Vasilevich A, Roumans N, Romero M, Dede Eren A, Tuvshindorj U, Alexander M, Carlier A, Williams P, Uquillas J, de Boer J. Expanding Biomaterial Surface Topographical Design Space through Natural Surface Reproduction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102084. [PMID: 34165820 PMCID: PMC11468538 DOI: 10.1002/adma.202102084] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/15/2021] [Indexed: 06/13/2023]
Abstract
Surface topography is a tool to endow biomaterials with bioactive properties. However, the large number of possible designs makes it challenging to find the optimal surface structure to induce a specific cell response. The TopoChip platform is currently the largest collection of topographies with 2176 in silico designed microtopographies. Still, it is exploring only a small part of the design space due to design algorithm limitations and the surface engineering strategy. Inspired by the diversity of natural surfaces, it is assessed as to what extent the topographical design space and consequently the resulting cellular responses can be expanded using natural surfaces. To this end, 26 plant and insect surfaces are replicated in polystyrene and their surface properties are quantified using white light interferometry. Through machine-learning algorithms, it is demonstrated that natural surfaces extend the design space of the TopoChip, which coincides with distinct morphological and focal adhesion profiles in mesenchymal stem cells (MSCs) and Pseudomonas aeruginosa colonization. Furthermore, differentiation experiments reveal the strong potential of the holy lotus to improve osteogenesis in MSCs. In the future, the design algorithms will be trained with the results obtained by natural surface imprint experiments to explore the bioactive properties of novel surface topographies.
Collapse
Affiliation(s)
- Steven Vermeulen
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Floris Honig
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Aliaksei Vasilevich
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Nadia Roumans
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Manuel Romero
- National Biofilms Innovation CentreBiodiscovery Institute and School of Life SciencesUniversity of NottinghamNottinghamNG7 2RDUK
| | - Aysegul Dede Eren
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Urnaa Tuvshindorj
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Morgan Alexander
- Advanced Materials and Healthcare TechnologiesThe School of PharmacyUniversity of NottinghamNottinghamNG7 2RDUK
| | - Aurélie Carlier
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Paul Williams
- National Biofilms Innovation CentreBiodiscovery Institute and School of Life SciencesUniversity of NottinghamNottinghamNG7 2RDUK
| | - Jorge Uquillas
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
10
|
Kerner J, Dogan A, von Recum H. Machine learning and big data provide crucial insight for future biomaterials discovery and research. Acta Biomater 2021; 130:54-65. [PMID: 34087445 DOI: 10.1016/j.actbio.2021.05.053] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Machine learning have been widely adopted in a variety of fields including engineering, science, and medicine revolutionizing how data is collected, used, and stored. Their implementation has led to a drastic increase in the number of computational models for the prediction of various numerical, categorical, or association events given input variables. We aim to examine recent advances in the use of machine learning when applied to the biomaterial field. Specifically, quantitative structure properties relationships offer the unique ability to correlate microscale molecular descriptors to larger macroscale material properties. These new models can be broken down further into four categories: regression, classification, association, and clustering. We examine recent approaches and new uses of machine learning in the three major categories of biomaterials: metals, polymers, and ceramics for rapid property prediction and trend identification. While current research is promising, limitations in the form of lack of standardized reporting and available databases complicates the implementation of described models. Herein, we hope to provide a snapshot of the current state of the field and a beginner's guide to navigating the intersection of biomaterials research and machine learning. STATEMENT OF SIGNIFICANCE: Machine learning and its methods have found a variety of uses beyond the field of computer science but have largely been neglected by those in realm of biomaterials. Through the use of more computational methods, biomaterials development can be expediated while reducing the need for standard trial and error methods. Within, we introduce four basic models that readers can potentially apply to their current research as well as current applications within the field. Furthermore, we hope that this article may act as a "call to action" for readers to realize and address the current lack of implementation within the biomaterials field.
Collapse
Affiliation(s)
- Jacob Kerner
- Case Western Reserve University; 10900 Euclid Ave., Cleveland Ohio 44106.
| | - Alan Dogan
- Case Western Reserve University; 10900 Euclid Ave., Cleveland Ohio 44106.
| | - Horst von Recum
- Case Western Reserve University; 10900 Euclid Ave., Cleveland Ohio 44106.
| |
Collapse
|
11
|
Soheilmoghaddam F, Rumble M, Cooper-White J. High-Throughput Routes to Biomaterials Discovery. Chem Rev 2021; 121:10792-10864. [PMID: 34213880 DOI: 10.1021/acs.chemrev.0c01026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many existing clinical treatments are limited in their ability to completely restore decreased or lost tissue and organ function, an unenviable situation only further exacerbated by a globally aging population. As a result, the demand for new medical interventions has increased substantially over the past 20 years, with the burgeoning fields of gene therapy, tissue engineering, and regenerative medicine showing promise to offer solutions for full repair or replacement of damaged or aging tissues. Success in these fields, however, inherently relies on biomaterials that are engendered with the ability to provide the necessary biological cues mimicking native extracellular matrixes that support cell fate. Accelerating the development of such "directive" biomaterials requires a shift in current design practices toward those that enable rapid synthesis and characterization of polymeric materials and the coupling of these processes with techniques that enable similarly rapid quantification and optimization of the interactions between these new material systems and target cells and tissues. This manuscript reviews recent advances in combinatorial and high-throughput (HT) technologies applied to polymeric biomaterial synthesis, fabrication, and chemical, physical, and biological screening with targeted end-point applications in the fields of gene therapy, tissue engineering, and regenerative medicine. Limitations of, and future opportunities for, the further application of these research tools and methodologies are also discussed.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Madeleine Rumble
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| |
Collapse
|
12
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
13
|
Karagöz Z, Rijns L, Dankers PY, van Griensven M, Carlier A. Towards understanding the messengers of extracellular space: Computational models of outside-in integrin reaction networks. Comput Struct Biotechnol J 2020; 19:303-314. [PMID: 33425258 PMCID: PMC7779863 DOI: 10.1016/j.csbj.2020.12.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The interactions between cells and their extracellular matrix (ECM) are critically important for homeostatic control of cell growth, proliferation, differentiation and apoptosis. Transmembrane integrin molecules facilitate the communication between ECM and the cell. Since the characterization of integrins in the late 1980s, there has been great advancement in understanding the function of integrins at different subcellular levels. However, the versatility in molecular pathways integrins are involved in, the high diversity in their interaction partners both outside and inside the cell as well as on the cell membrane and the short lifetime of events happening at the cell-ECM interface make it difficult to elucidate all the details regarding integrin function experimentally. To overcome the experimental challenges and advance the understanding of integrin biology, computational modeling tools have been used extensively. In this review, we summarize the computational models of integrin signaling while we explain the function of integrins at three main subcellular levels (outside the cell, cell membrane, cytosol). We also discuss how these computational modeling efforts can be helpful in other disciplines such as biomaterial design. As such, this review is a didactic modeling summary for biomaterial researchers interested in complementing their experimental work with computational tools or for seasoned computational scientists that would like to advance current in silico integrin models.
Collapse
Affiliation(s)
- Zeynep Karagöz
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Laura Rijns
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, the Netherlands
| | - Patricia Y.W. Dankers
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, the Netherlands
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
14
|
Vasilevich A, Carlier A, Winkler DA, Singh S, de Boer J. Evolutionary design of optimal surface topographies for biomaterials. Sci Rep 2020; 10:22160. [PMID: 33335124 PMCID: PMC7746696 DOI: 10.1038/s41598-020-78777-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 11/30/2020] [Indexed: 02/03/2023] Open
Abstract
Natural evolution tackles optimization by producing many genetic variants and exposing these variants to selective pressure, resulting in the survival of the fittest. We use high throughput screening of large libraries of materials with differing surface topographies to probe the interactions of implantable device coatings with cells and tissues. However, the vast size of possible parameter design space precludes a brute force approach to screening all topographical possibilities. Here, we took inspiration from Nature to optimize materials surface topographies using evolutionary algorithms. We show that successive cycles of material design, production, fitness assessment, selection, and mutation results in optimization of biomaterials designs. Starting from a small selection of topographically designed surfaces that upregulate expression of an osteogenic marker, we used genetic crossover and random mutagenesis to generate new generations of topographies.
Collapse
Affiliation(s)
- Aliaksei Vasilevich
- Institute for Complex Molecular Systems and Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - David A Winkler
- Materials Science & Engineering, Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia.,Monash Institute of Pharmaceutical Sciences, Monash Univeristy, Parkville, VIC, Australia.,Latrobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.,School of Pharmacy, University of Nottingham, Nottingham Park, UK
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jan de Boer
- Institute for Complex Molecular Systems and Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
15
|
Sujeeun LY, Goonoo N, Ramphul H, Chummun I, Gimié F, Baichoo S, Bhaw-Luximon A. Correlating in vitro performance with physico-chemical characteristics of nanofibrous scaffolds for skin tissue engineering using supervised machine learning algorithms. ROYAL SOCIETY OPEN SCIENCE 2020; 7:201293. [PMID: 33489277 PMCID: PMC7813265 DOI: 10.1098/rsos.201293] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/20/2020] [Indexed: 05/11/2023]
Abstract
The engineering of polymeric scaffolds for tissue regeneration has known a phenomenal growth during the past decades as materials scientists seek to understand cell biology and cell-material behaviour. Statistical methods are being applied to physico-chemical properties of polymeric scaffolds for tissue engineering (TE) to guide through the complexity of experimental conditions. We have attempted using experimental in vitro data and physico-chemical data of electrospun polymeric scaffolds, tested for skin TE, to model scaffold performance using machine learning (ML) approach. Fibre diameter, pore diameter, water contact angle and Young's modulus were used to find a correlation with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay of L929 fibroblasts cells on the scaffolds after 7 days. Six supervised learning algorithms were trained on the data using Seaborn/Scikit-learn Python libraries. After hyperparameter tuning, random forest regression yielded the highest accuracy of 62.74%. The predictive model was also correlated with in vivo data. This is a first preliminary study on ML methods for the prediction of cell-material interactions on nanofibrous scaffolds.
Collapse
Affiliation(s)
- Lakshmi Y. Sujeeun
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research (CBBR), University of Mauritius, 80837 Réduit, Mauritius
- Department of Digital Technologies, Faculty of Information, Communication and Digital Technologies, University of Mauritius, 80837 Réduit, Mauritius
| | - Nowsheen Goonoo
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research (CBBR), University of Mauritius, 80837 Réduit, Mauritius
| | - Honita Ramphul
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research (CBBR), University of Mauritius, 80837 Réduit, Mauritius
| | - Itisha Chummun
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research (CBBR), University of Mauritius, 80837 Réduit, Mauritius
| | - Fanny Gimié
- Animalerie, Plateforme de recherche CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, Ile de La Réunion, France
| | - Shakuntala Baichoo
- Department of Digital Technologies, Faculty of Information, Communication and Digital Technologies, University of Mauritius, 80837 Réduit, Mauritius
| | - Archana Bhaw-Luximon
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research (CBBR), University of Mauritius, 80837 Réduit, Mauritius
- Author for correspondence: Archana Bhaw-Luximon e-mail: ,
| |
Collapse
|
16
|
Isozaki A, Harmon J, Zhou Y, Li S, Nakagawa Y, Hayashi M, Mikami H, Lei C, Goda K. AI on a chip. LAB ON A CHIP 2020; 20:3074-3090. [PMID: 32644061 DOI: 10.1039/d0lc00521e] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Artificial intelligence (AI) has dramatically changed the landscape of science, industry, defence, and medicine in the last several years. Supported by considerably enhanced computational power and cloud storage, the field of AI has shifted from mostly theoretical studies in the discipline of computer science to diverse real-life applications such as drug design, material discovery, speech recognition, self-driving cars, advertising, finance, medical imaging, and astronomical observation, where AI-produced outcomes have been proven to be comparable or even superior to the performance of human experts. In these applications, what is essentially important for the development of AI is the data needed for machine learning. Despite its prominent importance, the very first process of the AI development, namely data collection and data preparation, is typically the most laborious task and is often a limiting factor of constructing functional AI algorithms. Lab-on-a-chip technology, in particular microfluidics, is a powerful platform for both the construction and implementation of AI in a large-scale, cost-effective, high-throughput, automated, and multiplexed manner, thereby overcoming the above bottleneck. On this platform, high-throughput imaging is a critical tool as it can generate high-content information (e.g., size, shape, structure, composition, interaction) of objects on a large scale. High-throughput imaging can also be paired with sorting and DNA/RNA sequencing to conduct a massive survey of phenotype-genotype relations whose data is too complex to analyze with traditional computational tools, but is analyzable with the power of AI. In addition to its function as a data provider, lab-on-a-chip technology can also be employed to implement the developed AI for accurate identification, characterization, classification, and prediction of objects in mixed, heterogeneous, or unknown samples. In this review article, motivated by the excellent synergy between AI and lab-on-a-chip technology, we outline fundamental elements, recent advances, future challenges, and emerging opportunities of AI with lab-on-a-chip technology or "AI on a chip" for short.
Collapse
Affiliation(s)
- Akihiro Isozaki
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and Kanagawa Institute of Industrial Science and Technology, Kanagawa 213-0012, Japan
| | - Jeffrey Harmon
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Yuqi Zhou
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Shuai Li
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and The Cambridge Centre for Data-Driven Discovery, Cambridge University, Cambridge CB3 0WA, UK
| | - Yuta Nakagawa
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Mika Hayashi
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Hideharu Mikami
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan.
| | - Cheng Lei
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and Institute of Technological Sciences, Wuhan University, Hubei 430072, China
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan. and Institute of Technological Sciences, Wuhan University, Hubei 430072, China and Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
17
|
Vermeulen S, de Boer J. Screening as a strategy to drive regenerative medicine research. Methods 2020; 190:80-95. [PMID: 32278807 DOI: 10.1016/j.ymeth.2020.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
In the field of regenerative medicine, optimization of the parameters leading to a desirable outcome remains a huge challenge. Examples include protocols for the guided differentiation of pluripotent cells towards specialized and functional cell types, phenotypic maintenance of primary cells in cell culture, or engineering of materials for improved tissue interaction with medical implants. This challenge originates from the enormous design space for biomaterials, chemical and biochemical compounds, and incomplete knowledge of the guiding biological principles. To tackle this challenge, high-throughput platforms allow screening of multiple perturbations in one experimental setup. In this review, we provide an overview of screening platforms that are used in regenerative medicine. We discuss their fabrication techniques, and in silico tools to analyze the extensive data sets typically generated by these platforms.
Collapse
Affiliation(s)
- Steven Vermeulen
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, the Netherlands; BioInterface Science Group, Department of Biomedical Engineering and Institute for Complex Molecular Systems, University of Eindhoven, Eindhoven, the Netherlands
| | - Jan de Boer
- BioInterface Science Group, Department of Biomedical Engineering and Institute for Complex Molecular Systems, University of Eindhoven, Eindhoven, the Netherlands.
| |
Collapse
|
18
|
Huang P, Guo YD, Zhang HW. Identification of Hub Genes in Pediatric Medulloblastoma by Multiple-Microarray Analysis. J Mol Neurosci 2019; 70:522-531. [DOI: 10.1007/s12031-019-01451-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
|
19
|
Dundas AA, Sanni O, Dubern JF, Dimitrakis G, Hook AL, Irvine DJ, Alexander PW, Alexander MR. Validating a Predictive Structure-Property Relationship by Discovery of Novel Polymers which Reduce Bacterial Biofilm Formation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1903513. [PMID: 31583791 PMCID: PMC7613244 DOI: 10.1002/adma.201903513] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/26/2019] [Indexed: 05/29/2023]
Abstract
Synthetic materials are an everyday component of modern healthcare yet often fail routinely as a consequence of medical-device-centered infections. The incidence rate for catheter-associated urinary tract infections is between 3% and 7% for each day of use, which means that infection is inevitable when resident for sufficient time. The O'Neill Review on antimicrobial resistance estimates that, left unchecked, ten million people will die annually from drug-resistant infections by 2050. Development of biomaterials resistant to bacterial colonization can play an important role in reducing device-associated infections. However, rational design of new biomaterials is hindered by the lack of quantitative structure-activity relationships (QSARs). Here, the development of a predictive QSAR is reported for bacterial biofilm formation on a range of polymers, using calculated molecular descriptors of monomer units to discover and exemplify novel, biofilm-resistant (meth-)acrylate-based polymers. These predictions are validated successfully by the synthesis of new monomers which are polymerized to create coatings found to be resistant to biofilm formation by six different bacterial pathogens: Pseudomonas aeruginosa, Proteus mirabilis, Enterococcus faecalis, Klebsiella pneumoniae, Escherichia coli, and Staphylococcus aureus.
Collapse
Affiliation(s)
- Adam A. Dundas
- Advanced Medical and Healthcare Technologies School of Pharmacy University of Nottingham Nottingham NG7 2RD, UK
| | - Olutoba Sanni
- Advanced Medical and Healthcare Technologies School of Pharmacy University of Nottingham Nottingham NG7 2RD, UK
- Department of Chemical and Environmental Engineering Faculty of Engineering University of Nottingham Nottingham NG7 2RD, UK
| | - Jean-Frédéric Dubern
- Centre of Biomolecular Sciences School of Life Sciences University of Nottingham Nottingham NG7 2RD, UK
| | - Georgios Dimitrakis
- Department of Chemical and Environmental Engineering Faculty of Engineering University of Nottingham Nottingham NG7 2RD, UK
| | - Andrew L. Hook
- Advanced Medical and Healthcare Technologies School of Pharmacy University of Nottingham Nottingham NG7 2RD, UK
| | - Derek J. Irvine
- Department of Chemical and Environmental Engineering Faculty of Engineering University of Nottingham Nottingham NG7 2RD, UK
| | - Paul Williams Alexander
- Centre of Biomolecular Sciences School of Life Sciences University of Nottingham Nottingham NG7 2RD, UK
| | | |
Collapse
|
20
|
Zimmermann JA, Schaffer DV. Engineering biomaterials to control the neural differentiation of stem cells. Brain Res Bull 2019; 150:50-60. [DOI: 10.1016/j.brainresbull.2019.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
|
21
|
Ashammakhi N, Darabi MA, Kehr NS, Erdem A, Hu SK, Dokmeci MR, Nasr AS, Khademhosseini A. Advances in Controlled Oxygen Generating Biomaterials for Tissue Engineering and Regenerative Therapy. Biomacromolecules 2019; 21:56-72. [DOI: 10.1021/acs.biomac.9b00546] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
| | - Mohammad Ali Darabi
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
| | - Nermin Seda Kehr
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Physikalisches Institut
and Center for Soft Nanoscience, Westfälische Wilhelms-Universität Münster, Busse-Peus-Strasse 10, 48149 Münster, Germany
| | - Ahmet Erdem
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry, Kocaeli University, Umuttepe Campus, 41380 Kocaeli, Turkey
- Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, 41380 Kocaeli, Turkey
| | - Shu-kai Hu
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Physikalisches Institut
and Center for Soft Nanoscience, Westfälische Wilhelms-Universität Münster, Busse-Peus-Strasse 10, 48149 Münster, Germany
| | - Mehmet R. Dokmeci
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
| | - Ali S. Nasr
- Division of Cardiothoracic Surgery, Department of Surgery, University of Iowa Hospitals and Clinics, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, United States
| | - Ali Khademhosseini
- Center for Minimally
Invasive Therapeutics (C-MIT), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Radiological Sciences, David Geffen School of Medicine, University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California−Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems
Institute (CNSI), University of California−Los Angeles, Los Angeles, California 90095, United States
- Department of Chemical Engineering, University of California−Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
22
|
Wieduwild R, Xu Y, Ostrovidov S, Khademhosseini A, Zhang Y, Orive G. Engineering Hydrogels beyond a Hydrated Network. Adv Healthc Mater 2019; 8:e1900038. [PMID: 30990968 DOI: 10.1002/adhm.201900038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/22/2019] [Indexed: 12/25/2022]
Abstract
In recent years, many mechanical, physical, chemical, and biochemical features of biomatrices have emerged as important properties to dictate the fates of cells. To construct chemically defined biomaterials to recapitulate various biological niches for both cell biology research and therapeutic utilities, it has become increasingly clear that a simple hydrated polymer network would not be able to provide the complex cues and signaling required for many types of cells. The researchers are facing a growing list of mechanophysical and biochemical properties, while each of them could be an important cellular trigger. To include all these design parameters in screening and synthesis is practically difficult, if not impossible. Developing novel high throughput screening technology by combining assay miniaturization, computer simulations, and modeling can help researchers to tackle the challenge to identify the most relevant parameters to tailor materials for specific applications.
Collapse
Affiliation(s)
- Robert Wieduwild
- Rudolf‐Schönheimer‐Institute of BiochemistryFaculty of MedicineLeipzig University Johannisallee 30 04103 Leipzig Germany
| | - Yong Xu
- B CUBE Center for Molecular BioengineeringTechnische Universität Dresden Tatzberg 41 01307 Dresden Germany
| | - Serge Ostrovidov
- Center for Minimally Invasive Therapeutics (C‐MIT) Los Angeles CA 90095 USA
- Department of Radiological SciencesUniversity of California ‐ Los Angeles Los Angeles CA 90095 USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C‐MIT) Los Angeles CA 90095 USA
- Department of BioengineeringUniversity of California ‐ Los Angeles Los Angeles CA 90095 USA
- Department of Radiological SciencesUniversity of California ‐ Los Angeles Los Angeles CA 90095 USA
- Department of Chemical and Biomolecular EngineeringUniversity of California ‐ Los Angeles Los Angeles CA 90095 USA
- California NanoSystems Institute (CNSI)University of California ‐ Los Angeles Los Angeles CA 90095 USA
| | - Yixin Zhang
- B CUBE Center for Molecular BioengineeringTechnische Universität Dresden Tatzberg 41 01307 Dresden Germany
| | - Gorka Orive
- NanoBioCel GroupLaboratory of PharmaceuticsSchool of PharmacyUniversity of the Basque Country UPV/EHU Paseo de la Universidad 7 01006 Vitoria‐Gasteiz Spain
- Biomedical Research Networking Centre in BioengineeringBiomaterials and Nanomedicine (CIBER‐BBN) Vitoria‐Gasteiz 01006 Spain
- University Institute for Regenerative Medicine and Oral Implantology ‐ UIRMI (UPV/EHU‐Fundación Eduardo Anitua) Vitoria 01007 Spain
- Singapore Eye Research InstituteThe Academia 20 College Road, Discovery Tower Singapore
| |
Collapse
|
23
|
Steier A, Muñiz A, Neale D, Lahann J. Emerging Trends in Information-Driven Engineering of Complex Biological Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806898. [PMID: 30957921 DOI: 10.1002/adma.201806898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/03/2018] [Indexed: 06/09/2023]
Abstract
Synthetic biological systems are used for a myriad of applications, including tissue engineered constructs for in vivo use and microengineered devices for in vitro testing. Recent advances in engineering complex biological systems have been fueled by opportunities arising from the combination of bioinspired materials with biological and computational tools. Driven by the availability of large datasets in the "omics" era of biology, the design of the next generation of tissue equivalents will have to integrate information from single-cell behavior to whole organ architecture. Herein, recent trends in combining multiscale processes to enable the design of the next generation of biomaterials are discussed. Any successful microprocessing pipeline must be able to integrate hierarchical sets of information to capture key aspects of functional tissue equivalents. Micro- and biofabrication techniques that facilitate hierarchical control as well as emerging polymer candidates used in these technologies are also reviewed.
Collapse
Affiliation(s)
- Anke Steier
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Ayşe Muñiz
- Biointerfaces Institute and Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dylan Neale
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joerg Lahann
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
- Biointerfaces Institute, Departments of Chemical Engineering, Materials Science and Engineering, and Biomedical Engineering and the, Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
24
|
Riordon J, Sovilj D, Sanner S, Sinton D, Young EW. Deep Learning with Microfluidics for Biotechnology. Trends Biotechnol 2019; 37:310-324. [DOI: 10.1016/j.tibtech.2018.08.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022]
|
25
|
Vasilevich A, de Boer J. Robot-scientists will lead tomorrow's biomaterials discovery. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018. [DOI: 10.1016/j.cobme.2018.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
26
|
Bouten CVC, Smits AIPM, Baaijens FPT. Can We Grow Valves Inside the Heart? Perspective on Material-based In Situ Heart Valve Tissue Engineering. Front Cardiovasc Med 2018; 5:54. [PMID: 29896481 PMCID: PMC5987128 DOI: 10.3389/fcvm.2018.00054] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022] Open
Abstract
In situ heart valve tissue engineering using cell-free synthetic, biodegradable scaffolds is under development as a clinically attractive approach to create living valves right inside the heart of a patient. In this approach, a valve-shaped porous scaffold "implant" is rapidly populated by endogenous cells that initiate neo-tissue formation in pace with scaffold degradation. While this may constitute a cost-effective procedure, compatible with regulatory and clinical standards worldwide, the new technology heavily relies on the development of advanced biomaterials, the processing thereof into (minimally invasive deliverable) scaffolds, and the interaction of such materials with endogenous cells and neo-tissue under hemodynamic conditions. Despite the first positive preclinical results and the initiation of a small-scale clinical trial by commercial parties, in situ tissue formation is not well understood. In addition, it remains to be determined whether the resulting neo-tissue can grow with the body and preserves functional homeostasis throughout life. More important yet, it is still unknown if and how in situ tissue formation can be controlled under conditions of genetic or acquired disease. Here, we discuss the recent advances of material-based in situ heart valve tissue engineering and highlight the most critical issues that remain before clinical application can be expected. We argue that a combination of basic science - unveiling the mechanisms of the human body to respond to the implanted biomaterial under (patho)physiological conditions - and technological advancements - relating to the development of next generation materials and the prediction of in situ tissue growth and adaptation - is essential to take the next step towards a realistic and rewarding translation of in situ heart valve tissue engineering.
Collapse
Affiliation(s)
- Carlijn V. C. Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Anthal I. P. M. Smits
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | - Frank P. T. Baaijens
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|