1
|
Hang R, Yao X, Bai L, Hang R. Evolving biomaterials design from trial and error to intelligent innovation. Acta Biomater 2025; 197:29-47. [PMID: 40081552 DOI: 10.1016/j.actbio.2025.03.013] [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: 11/20/2024] [Revised: 01/20/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The design and exploration of biomaterials plays a pivotal role in many fields, including medical and engineering. The prevailing approach to biomaterials discovery relies on orthogonal experiments, with repeated attempts to optimize experimental conditions. This method has proven invaluable in gaining experience, but it is also inefficient and challenging to predict the behavior of complex systems. The advent of high-throughput screening (HTS) techniques has led to a notable enhancement in the efficiency of biomaterials development, enabling researchers to assess a vast array of material combinations within a relatively short timeframe. Nevertheless, the emergence of artificial intelligence (AI) has been the catalyst for a new era in biomaterials design. AI has markedly accelerated the development of new materials by enabling the prediction of material properties through machine learning (ML) and deep learning models, as well as optimizing the design pipeline. This review will present a systematic overview of the development of biomaterials design technology. It will also explore the integration of AI with HTS technology and envisage the potential of AI-driven materials design in biomaterials for the future. STATEMENT OF SIGNIFICANCE: The design and synthesis of biomaterials have undergone substantial shifts, reflecting evolving research paradigms. High-throughput screening has emerged as a broad and efficient alternative to traditional free-form combination methods in biomaterial design. The advent of artificial intelligence (AI) enables personalized biomaterial design and, as a transformative tool in biomaterial development, is poised to redefine the field and offer long-term solutions for its advancement. Building on these advancements, this review systematically summarizes the evolution of biomaterial design, offering insights into the future trajectory of the field.
Collapse
Affiliation(s)
- Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, PR China.
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China.
| |
Collapse
|
2
|
Azuaje-Hualde E, Lartitegui-Meneses N, Alonso-Cabrera J, Inchaurraga-Llamas A, Alvarez-Braña Y, Martínez-dePancorbo M, Benito-Lopez F, Basabe-Desmonts L. CellStudio: a Modular, Tunable and Accessible Platform for Analysis of Growth Factors Secretions in Cell Cultures. ACS APPLIED MATERIALS & INTERFACES 2025; 17:8914-8923. [PMID: 39881577 DOI: 10.1021/acsami.4c17189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Traditional cell culture methods face significant limitations in monitoring cell secretions with spatial and temporal precision. Advanced microsystems incorporating biosensors have been developed to address these challenges, but they tend to lack versatility, and their complexity, along with the requirement for specialized equipment, limits their broader adoption. CellStudio offers an innovative, user-friendly solution that exploits Printing and Vacuum Lithography combined with bead-based assays to create modular and tunable cell patterns surrounded by biosensors. This platform allows for high-resolution, spatially resolved analysis of secreted proteins, such as VEGF and FGF-2, while being easily implementable in standard laboratory settings. CellStudio's design is compatible with conventional laboratory equipment, facilitating its integration into existing workflows without the need for extensive training or specialized tools. Validation experiments using mesenchymal stem cells and HeLa cells demonstrated that CellStudio can detect small secretion levels from small cell clusters with high sensitivity and analyze diffusion profiles, remarking the possibilities for studying cell behavior. By offering a standardized, cost-effective approach to detailed cellular analysis, CellStudio significantly enhances the capabilities of traditional cell culture techniques with broad applications across biological and biomedical research.
Collapse
Affiliation(s)
- Enrique Azuaje-Hualde
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Naiara Lartitegui-Meneses
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Juncal Alonso-Cabrera
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Asier Inchaurraga-Llamas
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Yara Alvarez-Braña
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Marian Martínez-dePancorbo
- BIOMICs Research Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Fernando Benito-Lopez
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
| | - Lourdes Basabe-Desmonts
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, University of the Basque Country UPV/EHU, Vitoria-Gasteiz 01006, Spain
- Basque Foundation of Science, IKERBASQUE, Bilbao 48011, Spain
| |
Collapse
|
3
|
Hang R, Zhao Y, Chen H, Li X, Yao R, Sun Y, Yao X, Bai L, Wang H, Han Y, Hang R. Construction and high-throughput screening of gradient nanowire coatings on titanium surface towards ameliorated osseointegration. Mater Today Bio 2025; 30:101392. [PMID: 39759850 PMCID: PMC11697249 DOI: 10.1016/j.mtbio.2024.101392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Surface nano-modification has emerged as an effective strategy to enhance osseointegration of titanium (Ti) implants. Despite its promise, rational optimization of surface nanomorphology for ameliorated osseointegration remains a significant challenge. Our research pioneering developed a one-step alkali etching technique to produce a gradient nanowire coating with continuously varied dimensions on Ti surfaces, which was subsequently served as a versatile platform for high-throughput screening of optimal dimensions to enhance osseointegration. The results showed that macrophages (MФs) that mainly governed the initial inflammatory reaction exhibited a polarization tendency towards pro-healing M2 phenotype with decreased nanowire dimension due to nanomorphology-mediated focal adhesion formation and activation of its downstream signaling pathways (typically PI3K-Akt). Simultaneously, small-sized nanowires with diameter of 5.63-14.25 nm and inter-spacing of 29.42-57.97 nm were conductive to angiogenesis of endothelial cells (ECs) and osteogenesis of bone marrow mesenchymal stem cells (BMSCs), which may share similar mechanisms of MФs. The in vivo results well corroborated these in vitro observations. The knowledge gained from the present work not only advance our understanding of the interaction between surface morphology and cells, but also potentially pave the way for efficient and cost-effective design of advanced biomaterial surfaces for better osseointegration.
Collapse
Affiliation(s)
- Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Yuyu Zhao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Huanming Chen
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Xiaomei Li
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030012, China
| | - Runhua Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Yonghua Sun
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yong Han
- State-Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, 030024, China
- State-Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
4
|
Seymour S, Cadena I, Johnson M, Thakkar R, Jenne M, Adem I, Almer A, Frankovic R, Spence D, Haddadin A, Fogg KC. Empowering High Throughput Screening of 3D Models: Automated Dispensing of Cervical and Endometrial Cancer Cells. Cell Mol Bioeng 2025; 18:71-82. [PMID: 39949489 PMCID: PMC11813830 DOI: 10.1007/s12195-024-00841-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 12/17/2024] [Indexed: 02/16/2025] Open
Abstract
Purpose Cervical and endometrial cancers pose significant challenges in women's healthcare due to their high mortality rates and limited treatment options. High throughput screening (HTS) of cervical and endometrial cancer in vitro models offers a promising avenue for drug repurposing and broadening patient treatment options. Traditional two-dimensional (2D) cell-based screenings have limited capabilities to capture crucial multicellular interactions, that are improved upon in three dimensional (3D) multicellular tissue engineered models. However, manual fabrication of the 3D platforms is both time consuming and subject to variability. Thus, the goal of this study was to utilize automated cell dispensing to fabricate 3D cell-based HTS platforms using the HP D100 Single Cell Dispenser to dispense cervical and endometrial cancer cells. Methods We evaluated the effects of automated dispensing of the cancer cell lines by tuning the dispensing protocol to align with cell size measured in solution and the minimum cell number for acceptable cell viability and proliferation. We modified our previously reported coculture models of cervical and endometrial cancer to be in a 384 well plate format and measured microvessel length and cancer cell invasion. Results Automatically and manually dispensed cells were directly compared revealing minimal differences between the dispensing methods. These findings suggest that automated dispensing of cancer cells minimally affects cell behavior and can be deployed to decrease in vitro model fabrication time. Conclusions By streamlining the manufacturing process, automated dispensing holds promise for enhancing efficiency and scalability of 3D in vitro HTS platforms, ultimately contributing to advancement in cancer research and treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00841-y.
Collapse
Affiliation(s)
- Samantha Seymour
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Ines Cadena
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | | | - Riya Thakkar
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Molly Jenne
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Iman Adem
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Alyssa Almer
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Rachael Frankovic
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Danielle Spence
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Andrea Haddadin
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| | - Kaitlin C. Fogg
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, OR 97330 USA
| |
Collapse
|
5
|
Azuaje-Hualde E, Alonso-Cabrera JA, de Pancorbo MM, Benito-Lopez F, Basabe-Desmonts L. Integration of secreted signaling molecule sensing on cell monitoring platforms: a critical review. Anal Bioanal Chem 2024; 416:7249-7266. [PMID: 39048740 PMCID: PMC11584473 DOI: 10.1007/s00216-024-05435-1] [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: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
Monitoring cell secretion in complex microenvironments is crucial for understanding cellular behavior and advancing physiological and pathological research. While traditional cell culture methods, including organoids and spheroids, provide valuable models, real-time monitoring of cell secretion of signaling molecules remains challenging. Integrating advanced monitoring technologies into these systems often disrupts the delicate balance of the microenvironment, making it difficult to achieve sensitivity and specificity. This review explored recent strategies for integrating the monitoring of cell secretion of signaling molecules, crucial for understanding and replicating cell microenvironments, within cell culture platforms, addressing challenges such as non-adherent cell models and the focus on single-cell methodologies. We highlight advancements in biosensors, microfluidics, and three-dimensional culture methods, and discuss their potential to enhance real-time, multiplexed cell monitoring. By examining the advantages, limitations, and future prospects of these technologies, we aim to contribute to the development of integrated systems that facilitate comprehensive cell monitoring, ultimately advancing biological research and pharmaceutical development.
Collapse
Affiliation(s)
- Enrique Azuaje-Hualde
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Juncal A Alonso-Cabrera
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Fernando Benito-Lopez
- Microfluidics Cluster UPV/EHU, Analytical Microsystems & Materials for Lab-on-a-Chip (AMMa-LOAC) Group, Analytical Chemistry Department, University of the Basque Country UPV/EHU, Leioa, Spain.
- Microfluidics Cluster UPV/EHU, Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
- Basque Foundation of Science, IKERBASQUE, María Díaz Haroko Kalea, 3, 48013, Bilbao, Spain.
| | - Lourdes Basabe-Desmonts
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.
- Microfluidics Cluster UPV/EHU, Bioaraba Health Research Institute, Vitoria-Gasteiz, Spain.
- Basque Foundation of Science, IKERBASQUE, María Díaz Haroko Kalea, 3, 48013, Bilbao, Spain.
| |
Collapse
|
6
|
Miller RC, Temenoff JS. Biomaterials for Cell Manufacturing. ACS Macro Lett 2024; 13:1521-1530. [PMID: 39466845 PMCID: PMC11580378 DOI: 10.1021/acsmacrolett.4c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Cell therapies, potent populations of cells used to treat disease and injury, can be strategically manufactured with biomaterial intervention to improve clinical translation. In this viewpoint, we discuss biomaterial design and integration into cell manufacturing steps to achieve three main goals: scale-up, phenotype control, and selection of potent cells. Material properties can be engineered to influence the cell-biomaterial interface and, therefore, impart desirable cell behavior such as growth, secretory activity, and differentiation. Future directions for the field should capitalize on the combinatorial design of biomaterial properties to yield highly specific and potent cell populations. Furthermore, future biomaterials could contribute to novel high-throughput cell separation technologies that can individually select the most therapeutically relevant cells within a produced batch.
Collapse
Affiliation(s)
- Ryan C. Miller
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech/Emory University, Atlanta, Georgia 30332, United States
| | - Johnna S. Temenoff
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech/Emory University, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
7
|
Singh A, Cho YK, Cohen DJ. Rapid Whole-Plate Cell and Tissue Micropatterning Using a Budget 3D Resin Printer. ACS OMEGA 2024; 9:43808-43816. [PMID: 39494000 PMCID: PMC11525498 DOI: 10.1021/acsomega.4c06539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024]
Abstract
The ability to precisely pattern cells and proteins is crucial in various scientific disciplines, including cell biology, bioengineering, and materials chemistry. Current techniques, such as microcontact stamping, 3D bioprinting, and direct photopatterning, have limitations in terms of cost, versatility, and throughput. In this Article, we present an accessible approach that combines the throughput of photomask systems with the versatility of programmable light patterning using a low-cost consumer LCD resin printer. The method involves utilizing a bioinert hydrogel, poly(ethylene glycol) diacrylate (PEGDA), and a 405 nm sensitive photoinitiator (LAP) that are selectively cross-linked to form a hydrogel upon light exposure, creating specific regions that are protein and cell-repellent. Our result highlights that a low-cost LCD resin printer can project virtual photomasks onto the hydrogel, allowing for reasonable resolution and large-area printing at a fraction of the cost of traditional systems. The study demonstrates the calibration of exposure times for optimal resolution and accuracy and shape corrections to overcome the inherent challenges of wide-field resin printing. The potential of this approach is validated through widely studied 2D and 3D stem cell applications, showcasing its biocompatibility and ability to replicate complex tissue engineering patterns. We also validate the method with a cell-adhesive polymer (gelatin methacrylate; GelMA). The combination of low cost, high throughput, and accessibility makes this method broadly applicable across fields for enabling rapid and precise fabrication of cells and tissues in standard laboratory culture vessels.
Collapse
Affiliation(s)
- Anamika Singh
- Department
of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Youn Kyoung Cho
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Daniel J. Cohen
- Department
of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
8
|
Ku J, Asuri P. Stem cell-based approaches for developmental neurotoxicity testing. FRONTIERS IN TOXICOLOGY 2024; 6:1402630. [PMID: 39238878 PMCID: PMC11374538 DOI: 10.3389/ftox.2024.1402630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Neurotoxicants are substances that can lead to adverse structural or functional effects on the nervous system. These can be chemical, biological, or physical agents that can cross the blood brain barrier to damage neurons or interfere with complex interactions between the nervous system and other organs. With concerns regarding social policy, public health, and medicine, there is a need to ensure rigorous testing for neurotoxicity. While the most common neurotoxicity tests involve using animal models, a shift towards stem cell-based platforms can potentially provide a more biologically accurate alternative in both clinical and pharmaceutical research. With this in mind, the objective of this article is to review both current technologies and recent advancements in evaluating neurotoxicants using stem cell-based approaches, with an emphasis on developmental neurotoxicants (DNTs) as these have the most potential to lead to irreversible critical damage on brain function. In the next section, attempts to develop novel predictive model approaches for the study of both neural cell fate and developmental neurotoxicity are discussed. Finally, this article concludes with a discussion of the future use of in silico methods within developmental neurotoxicity testing, and the role of regulatory bodies in promoting advancements within the space.
Collapse
Affiliation(s)
- Joy Ku
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, United States
| | - Prashanth Asuri
- Department of Bioengineering, Santa Clara University, Santa Clara, CA, United States
| |
Collapse
|
9
|
Sytu MRC, Cho DH, Hahm JI. Self-Assembled Block Copolymers as a Facile Pathway to Create Functional Nanobiosensor and Nanobiomaterial Surfaces. Polymers (Basel) 2024; 16:1267. [PMID: 38732737 PMCID: PMC11085100 DOI: 10.3390/polym16091267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Block copolymer (BCP) surfaces permit an exquisite level of nanoscale control in biomolecular assemblies solely based on self-assembly. Owing to this, BCP-based biomolecular assembly represents a much-needed, new paradigm for creating nanobiosensors and nanobiomaterials without the need for costly and time-consuming fabrication steps. Research endeavors in the BCP nanobiotechnology field have led to stimulating results that can promote our current understanding of biomolecular interactions at a solid interface to the never-explored size regimes comparable to individual biomolecules. Encouraging research outcomes have also been reported for the stability and activity of biomolecules bound on BCP thin film surfaces. A wide range of single and multicomponent biomolecules and BCP systems has been assessed to substantiate the potential utility in practical applications as next-generation nanobiosensors, nanobiodevices, and biomaterials. To this end, this Review highlights pioneering research efforts made in the BCP nanobiotechnology area. The discussions will be focused on those works particularly pertaining to nanoscale surface assembly of functional biomolecules, biomolecular interaction properties unique to nanoscale polymer interfaces, functionality of nanoscale surface-bound biomolecules, and specific examples in biosensing. Systems involving the incorporation of biomolecules as one of the blocks in BCPs, i.e., DNA-BCP hybrids, protein-BCP conjugates, and isolated BCP micelles of bioligand carriers used in drug delivery, are outside of the scope of this Review. Looking ahead, there awaits plenty of exciting research opportunities to advance the research field of BCP nanobiotechnology by capitalizing on the fundamental groundwork laid so far for the biomolecular interactions on BCP surfaces. In order to better guide the path forward, key fundamental questions yet to be addressed by the field are identified. In addition, future research directions of BCP nanobiotechnology are contemplated in the concluding section of this Review.
Collapse
Affiliation(s)
- Marion Ryan C. Sytu
- Department of Chemistry, Georgetown University, 37th & O Sts. NW., Washington, DC 20057, USA
| | - David H. Cho
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA;
| | - Jong-in Hahm
- Department of Chemistry, Georgetown University, 37th & O Sts. NW., Washington, DC 20057, USA
| |
Collapse
|
10
|
Nao SC, Huang LS, Shiu-Hin Chan D, Wang X, Li GD, Wu J, Wong CY, Wang W, Leung CH. Repurposing sodium stibogluconate as an uracil DNA glycosylase inhibitor against prostate cancer using a time-resolved oligonucleotide-based drug screening platform. Bioorg Chem 2024; 144:107176. [PMID: 38330721 DOI: 10.1016/j.bioorg.2024.107176] [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] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024]
Abstract
Repurposing drugs can significantly reduce the time and costs associated with drug discovery and development. However, many drug compounds possess intrinsic fluorescence, resulting in aberrations such as auto-fluorescence, scattering and quenching, in fluorescent high-throughput screening assays. To overcome these drawbacks, time-resolved technologies have received increasing attention. In this study, we have developed a rapid and efficient screening platform based on time-resolved emission spectroscopy in order to screen for inhibitors of the DNA repair enzyme, uracil-DNA glycosylase (UDG). From a database of 1456 FDA/EMA-approved drugs, sodium stibogluconate was discovered as a potent UDG inhibitor. This compound showed synergistic cytotoxicity against 5-fluorouracil-resistant cancer cells. This work provides a promising future for time-resolved technologies for high-throughput screening (HTS), allowing for the swift identification of bioactive compounds from previously overlooked scaffolds due to their inherent fluorescence properties.
Collapse
Affiliation(s)
- Sang-Cuo Nao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Le-Sheng Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | | | - Xueliang Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China
| | - Guo-Dong Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chun-Yuen Wong
- Department of Chemistry, City University of Hong Kong, Hong Kong, China.
| | - Wanhe Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen 518057, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Taipa, Macau, China; MoE Frontiers Science Centre for Precision Oncology, University of Macau, Taipa, Macau, China.
| |
Collapse
|
11
|
Yang Z, Chen J, Xiao Y, Yang C, Zhao CX, Chen D, Weitz DA. Digital Barcodes for High-Throughput Screening. CHEM & BIO ENGINEERING 2024; 1:2-12. [PMID: 39973970 PMCID: PMC11835184 DOI: 10.1021/cbe.3c00085] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2025]
Abstract
High-throughput screening is an indispensable technology in drug discovery, cancer therapy, and disease diagnosis, and it could greatly reduce time cost, reagent consumption, and labor expense. Here, four high-throughput screening methods with high sensitivity and accessibility are discussed in detail. Fluorescence, DNA, heavy metal, and nonmetal isotope barcodes, which generally label antibodies, proteins, and saccharides to identify cells, are detected by flow cytometry, second-generation DNA sequencing, mass cytometry, and second-ion mass spectrometry, respectively. Encoding binary information in barcodes, labeling individual cells by barcodes, performing the characterization of cells together, and identifying the result belonging to individual cells via barcodes are the main steps for high-throughput screening. Applications of the four digital barcodes in high-throughput screening for both in vitro and in vivo tests are described in detail, and their advantages and disadvantages are also summarized. High-throughput screening has provided a powerful platform widely accessible for multidisciplinary studies and has greatly sped up the progress of drug discovery, disease diagnosis, and cancer therapy.
Collapse
Affiliation(s)
- Ze Yang
- College
of Energy Engineering and State Key Laboratory of Clean Energy Utilization, Zhejiang University, Hangzhou 310003, Zhejiang
Province, People’s Republic
of China
- Zhejiang
Key Laboratory of Smart Biomaterials, College of Chemical and Biological
Engineering, Zhejiang University, Hangzhou 310027, Zhejiang Province, People’s Republic of China
| | - Jingyi Chen
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Yao Xiao
- College
of Energy Engineering and State Key Laboratory of Clean Energy Utilization, Zhejiang University, Hangzhou 310003, Zhejiang
Province, People’s Republic
of China
| | - Chenjing Yang
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
- Wenzhou
Institute, University of Chinese Academy
of Sciences, Wenzhou, Zhejiang 325001, People’s Republic of China
| | - Chun-Xia Zhao
- School
of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Dong Chen
- College
of Energy Engineering and State Key Laboratory of Clean Energy Utilization, Zhejiang University, Hangzhou 310003, Zhejiang
Province, People’s Republic
of China
- Zhejiang
Key Laboratory of Smart Biomaterials, College of Chemical and Biological
Engineering, Zhejiang University, Hangzhou 310027, Zhejiang Province, People’s Republic of China
- Department
of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang
Province, People’s Republic
of China
| | - David A. Weitz
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
12
|
Mahdi-Esferizi R, Haji Molla Hoseyni B, Mehrpanah A, Golzade Y, Najafi A, Elahian F, Zadeh Shirazi A, Gomez GA, Tahmasebian S. DeeP4med: deep learning for P4 medicine to predict normal and cancer transcriptome in multiple human tissues. BMC Bioinformatics 2023; 24:275. [PMID: 37403016 DOI: 10.1186/s12859-023-05400-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/25/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND P4 medicine (predict, prevent, personalize, and participate) is a new approach to diagnosing and predicting diseases on a patient-by-patient basis. For the prevention and treatment of diseases, prediction plays a fundamental role. One of the intelligent strategies is the design of deep learning models that can predict the state of the disease using gene expression data. RESULTS We create an autoencoder deep learning model called DeeP4med, including a Classifier and a Transferor that predicts cancer's gene expression (mRNA) matrix from its matched normal sample and vice versa. The range of the F1 score of the model, depending on tissue type in the Classifier, is from 0.935 to 0.999 and in Transferor from 0.944 to 0.999. The accuracy of DeeP4med for tissue and disease classification was 0.986 and 0.992, respectively, which performed better compared to seven classic machine learning models (Support Vector Classifier, Logistic Regression, Linear Discriminant Analysis, Naive Bayes, Decision Tree, Random Forest, K Nearest Neighbors). CONCLUSIONS Based on the idea of DeeP4med, by having the gene expression matrix of a normal tissue, we can predict its tumor gene expression matrix and, in this way, find effective genes in transforming a normal tissue into a tumor tissue. Results of Differentially Expressed Genes (DEGs) and enrichment analysis on the predicted matrices for 13 types of cancer showed a good correlation with the literature and biological databases. This led that by using the gene expression matrix, to train the model with features of each person in a normal and cancer state, this model could predict diagnosis based on gene expression data from healthy tissue and be used to identify possible therapeutic interventions for those patients.
Collapse
Affiliation(s)
- Roohallah Mahdi-Esferizi
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Amir Mehrpanah
- Faculty of Mathematics, Shahid Beheshti University, Tehran, Iran
| | - Yazdan Golzade
- Department of Mathematics, Faculty of Basic Sciences, Iran University of Science and Technology,(IUST), Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Fatemeh Elahian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Zadeh Shirazi
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia
| | - Guillermo A Gomez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, 5000, Australia
| | - Shahram Tahmasebian
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
13
|
Dong J, Wang W, Zhou W, Zhang S, Li M, Li N, Pan G, Zhang X, Bai J, Zhu C. Immunomodulatory biomaterials for implant-associated infections: from conventional to advanced therapeutic strategies. Biomater Res 2022; 26:72. [PMID: 36471454 PMCID: PMC9721013 DOI: 10.1186/s40824-022-00326-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/19/2022] [Indexed: 12/11/2022] Open
Abstract
Implant-associated infection (IAI) is increasingly emerging as a serious threat with the massive application of biomaterials. Bacteria attached to the surface of implants are often difficult to remove and exhibit high resistance to bactericides. In the quest for novel antimicrobial strategies, conventional antimicrobial materials often fail to exert their function because they tend to focus on direct bactericidal activity while neglecting the modulation of immune systems. The inflammatory response induced by host immune cells was thought to be a detrimental force impeding wound healing. However, the immune system has recently received increasing attention as a vital player in the host's defense against infection. Anti-infective strategies based on the modulation of host immune defenses are emerging as a field of interest. This review explains the importance of the immune system in combating infections and describes current advanced immune-enhanced anti-infection strategies. First, the characteristics of traditional/conventional implant biomaterials and the reasons for the difficulty of bacterial clearance in IAI were reviewed. Second, the importance of immune cells in the battle against bacteria is elucidated. Then, we discuss how to design biomaterials that activate the defense function of immune cells to enhance the antimicrobial potential. Based on the key premise of restoring proper host-protective immunity, varying advanced immune-enhanced antimicrobial strategies were discussed. Finally, current issues and perspectives in this field were offered. This review will provide scientific guidance to enhance the development of advanced anti-infective biomaterials.
Collapse
Affiliation(s)
- Jiale Dong
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Wenzhi Wang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Wei Zhou
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Siming Zhang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Meng Li
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China ,grid.263761.70000 0001 0198 0694Medical College, Soochow University, 215006 Suzhou, Jiangsu P. R. China
| | - Ning Li
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Guoqing Pan
- grid.440785.a0000 0001 0743 511XInstitute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 212013 Zhenjiang, China
| | - Xianzuo Zhang
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| | - Jiaxiang Bai
- grid.263761.70000 0001 0198 0694Medical College, Soochow University, 215006 Suzhou, Jiangsu P. R. China
| | - Chen Zhu
- grid.411395.b0000 0004 1757 0085Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, 230001 Hefei, Anhui P. R. China
| |
Collapse
|
14
|
Zhao H, Chen T, Wu T, Xie L, Ma Y, Sha J. Strategy based on multiplexed brush architectures for regulating the spatiotemporal immobilization of biomolecules. BIOMATERIALS ADVANCES 2022; 141:213092. [PMID: 36191539 DOI: 10.1016/j.bioadv.2022.213092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/03/2022] [Accepted: 08/20/2022] [Indexed: 06/16/2023]
Abstract
Functional surfaces that enable both spatial and temporal control of biomolecules immobilization have attracted enormous attention for various fields including smart biointerface materials, high-throughput bioarrays, and fundamental research in the biosciences. Here, a flexible and promising method was presented for regulating the spatiotemporal arrangement of multiple biomolecules by constructing the topographically and chemically diverse polymer brushes patterned surfaces. A series of polymer brushes patterned surfaces, including antifouling brushes patterned surface, epoxy-presenting brushes patterned surface without and with antifouling background layer, were fabricated to control the spatial distribution of protein and cell adhesion through specific and nonspecific means. The fluorescence measurements demonstrated the effectiveness of spatially regulating the density of surface-immobilized protein through controlling the areal thickness of the poly (glycidyl methacrylate) (PGMA) brush patterns, leading to various complex patterns featuring well-defined biomolecule concentration gradients. Furthermore, a multiplexed surface bearing epoxy groups and azido groups with various areal densities was fabricated for regulating the spatiotemporal arrangement of different proteins, enabling binary biomolecules patterns with higher degrees of functionality and complexity. The presented strategy for the spatiotemporal control of biomolecules immobilization would boost the development of dynamic and multifunctional biosystems.
Collapse
Affiliation(s)
- Haili Zhao
- Faculty of Chemical Engineering, Kunming University of Science and Technology, Kunming 650504, China
| | - Tao Chen
- Faculty of Chemical Engineering, Kunming University of Science and Technology, Kunming 650504, China
| | - Tong Wu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Linsheng Xie
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yulu Ma
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jin Sha
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
15
|
Yang L, Conley BM, Yoon J, Rathnam C, Pongkulapa T, Conklin B, Hou Y, Lee KB. High-Content Screening and Analysis of Stem Cell-Derived Neural Interfaces Using a Combinatorial Nanotechnology and Machine Learning Approach. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9784273. [PMID: 36204248 PMCID: PMC9513834 DOI: 10.34133/2022/9784273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022]
Abstract
A systematic investigation of stem cell-derived neural interfaces can facilitate the discovery of the molecular mechanisms behind cell behavior in neurological disorders and accelerate the development of stem cell-based therapies. Nevertheless, high-throughput investigation of the cell-type-specific biophysical cues associated with stem cell-derived neural interfaces continues to be a significant obstacle to overcome. To this end, we developed a combinatorial nanoarray-based method for high-throughput investigation of neural interface micro-/nanostructures (physical cues comprising geometrical, topographical, and mechanical aspects) and the effects of these complex physical cues on stem cell fate decisions. Furthermore, by applying a machine learning (ML)-based analytical approach to a large number of stem cell-derived neural interfaces, we comprehensively mapped stem cell adhesion, differentiation, and proliferation, which allowed for the cell-type-specific design of biomaterials for neural interfacing, including both adult and human-induced pluripotent stem cells (hiPSCs) with varying genetic backgrounds. In short, we successfully demonstrated how an innovative combinatorial nanoarray and ML-based platform technology can aid with the rational design of stem cell-derived neural interfaces, potentially facilitating precision, and personalized tissue engineering applications.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Brian M. Conley
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jinho Yoon
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
16
|
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
|
17
|
Chen Y, Sun W, Tang H, Li Y, Li C, Wang L, Chen J, Lin W, Li S, Fan Z, Cheng Y, Chen C. Interactions Between Immunomodulatory Biomaterials and Immune Microenvironment: Cues for Immunomodulation Strategies in Tissue Repair. Front Bioeng Biotechnol 2022; 10:820940. [PMID: 35646833 PMCID: PMC9140325 DOI: 10.3389/fbioe.2022.820940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The foreign body response (FBR) caused by biomaterials can essentially be understood as the interaction between the immune microenvironment and biomaterials, which has severely impeded the application of biomaterials in tissue repair. This concrete interaction occurs via cells and bioactive substances, such as proteins and nucleic acids. These cellular and molecular interactions provide important cues for determining which element to incorporate into immunomodulatory biomaterials (IMBs), and IMBs can thus be endowed with the ability to modulate the FBR and repair damaged tissue. In terms of cellular, IMBs are modified to modulate functions of immune cells, such as macrophages and mast cells. In terms of bioactive substances, proteins and nucleic acids are delivered to influence the immune microenvironment. Meanwhile, IMBs are designed with high affinity for spatial targets and the ability to self-adapt over time, which allows for more efficient and intelligent tissue repair. Hence, IMB may achieve the perfect functional integration in the host, representing a breakthrough in tissue repair and regeneration medicine.
Collapse
Affiliation(s)
- Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yingze Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Li
- School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Shenghui Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Ziwen Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yu Cheng
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| |
Collapse
|
18
|
Park SJ, Jung TH, Kim JH, Lee KY, Kim J, Ju J, Moon SH. In silico design and fabrication of an SFI chip-based microspheroid culture system. Biomater Sci 2022; 10:2991-3005. [PMID: 35521942 DOI: 10.1039/d2bm00250g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The emergence of microfluidic devices and computational fluid dynamics (CFD) has propelled the need for next-generation biomimetic cell culture platforms that are flexible for monitoring and regulation. Therefore, this study evaluated a CFD application in an in silico-designed and spheroid-based flow integration 3D cell culture chip (SFI chip) to illustrate cell culture, drug screening, cytokine delivery, and differentiation of cells in a platform that partially recapitulates the natural environment. Our results show that a flow rate of 0.05 mL h-1 or less induced no physical stress in the SFI chip (15 mm), and uniform cell spheroids (approximately 200 μm) were formed across the platform. The cultured cells were tested in several experimental contexts (co-culture, drug screening, cytokine delivery, and differentiation), demonstrating the usefulness of computational simulation in expediting discovery and simple and effective means to scale the production of standardized cell spheroids cultured under dynamic and natural conditions. Advanced cell culture technologies can be used to accelerate research and discovery and the preclinical and clinical development of cell and cell-free therapies for urgent medical needs.
Collapse
Affiliation(s)
- Soon-Jung Park
- Department of Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea.,Stem Cell Research Institute, T&R Biofab Co. Ltd, Siheung, Republic of Korea.
| | - Taek-Hee Jung
- Department of Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea.,Stem Cell Research Institute, T&R Biofab Co. Ltd, Siheung, Republic of Korea.
| | - Jong Hyun Kim
- Department of Biological Science, Hyupsung University, Hwasung, Republic of Korea
| | - Kyoung-Yong Lee
- Carbon Neutral Technology R&D Department, Korea Institute of Industrial Technology (KITECH), Cheonan, Republic of Korea
| | - Jeongyun Kim
- Department of Physics, College of Science & Technology, Dankook University, Cheonan, Chungnam, 31116, Republic of Korea.
| | - Jongil Ju
- Department of Physics, College of Science & Technology, Dankook University, Cheonan, Chungnam, 31116, Republic of Korea. .,Department of R&D, ABM Scientific Co., Cheonan, Republic of Korea
| | - Sung-Hwan Moon
- Department of Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea.,Stem Cell Research Institute, T&R Biofab Co. Ltd, Siheung, Republic of Korea. .,Department of Animal Biotechnology, Sangji University, Wonju, Republic of Korea
| |
Collapse
|
19
|
Yang L, Conley BM, Rathnam C, Cho HY, Pongkulapa T, Conklin B, Lee KB. Predictive Biophysical Cue Mapping for Direct Cell Reprogramming Using Combinatorial Nanoarrays. ACS NANO 2022; 16:5577-5586. [PMID: 35301847 DOI: 10.1021/acsnano.1c10344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biophysical cues, such as nanotopographies of extracellular matrix (ECM), are key cell regulators for direct cell reprogramming. Therefore, high-throughput methods capable of systematically screening a wide range of biophysical cue-regulated cell reprogramming are increasingly needed for tissue engineering and regenerative medicine. Here, we report the development of a dynamic laser interference lithography (DIL) to generate large-scale combinatorial biophysical cue (CBC) arrays with diverse micro/nanostructures at higher complexities than most current arrays. Using CBC arrays, a high-throughput cell mapping method is further demonstrated for the systematic investigation of biophysical cue-mediated direct cell reprogramming. This CBC array-based high-throughput cell screening approach facilitates the rapid identification of unconventional hierarchical nanopatterns that induce the direct reprogramming of human fibroblasts into neurons through epigenetic modulation mechanisms. In this way, we successfully demonstrate DIL for generating highly complex CBC arrays and establish CBC array-based cell screening as a valuable strategy for systematically investigating the role of biophysical cues in cell reprogramming.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Brian M Conley
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Hyeon-Yeol Cho
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| |
Collapse
|
20
|
Bonini F, Mosser S, Mor FM, Boutabla A, Burch P, Béduer A, Roux A, Braschler T. The Role of Interstitial Fluid Pressure in Cerebral Porous Biomaterial Integration. Brain Sci 2022; 12:417. [PMID: 35447953 PMCID: PMC9040716 DOI: 10.3390/brainsci12040417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 02/05/2023] Open
Abstract
Recent advances in biomaterials offer new possibilities for brain tissue reconstruction. Biocompatibility, provision of cell adhesion motives and mechanical properties are among the present main design criteria. We here propose a radically new and potentially major element determining biointegration of porous biomaterials: the favorable effect of interstitial fluid pressure (IFP). The force applied by the lymphatic system through the interstitial fluid pressure on biomaterial integration has mostly been neglected so far. We hypothesize it has the potential to force 3D biointegration of porous biomaterials. In this study, we develop a capillary hydrostatic device to apply controlled in vitro interstitial fluid pressure and study its effect during 3D tissue culture. We find that the IFP is a key player in porous biomaterial tissue integration, at physiological IFP levels, surpassing the known effect of cell adhesion motives. Spontaneous electrical activity indicates that the culture conditions are not harmful for the cells. Our work identifies interstitial fluid pressure at physiological negative values as a potential main driver for tissue integration into porous biomaterials. We anticipate that controlling the IFP level could narrow the gap between in vivo and in vitro and therefore decrease the need for animal screening in biomaterial design.
Collapse
Affiliation(s)
- Fabien Bonini
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
| | - Sébastien Mosser
- Neurix SA, Avenue de la Roseraie 64, CH-1022 Geneva, Switzerland;
| | - Flavio Maurizio Mor
- Haute École du Paysage, d’Ingénierie et d’Architecture de Genève, Haute École Spécialisée de Suisse Occidentale (HEPIA HES-SO), University of Applied Sciences and Arts Western Switzerland, CH-1202 Geneva, Switzerland; (F.M.M.); (A.R.)
| | - Anissa Boutabla
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
| | - Patrick Burch
- Volumina-Medical SA, Route de la Corniche 5, CH-1066 Epalinges, Switzerland;
| | - Amélie Béduer
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
- Volumina-Medical SA, Route de la Corniche 5, CH-1066 Epalinges, Switzerland;
| | - Adrien Roux
- Haute École du Paysage, d’Ingénierie et d’Architecture de Genève, Haute École Spécialisée de Suisse Occidentale (HEPIA HES-SO), University of Applied Sciences and Arts Western Switzerland, CH-1202 Geneva, Switzerland; (F.M.M.); (A.R.)
| | - Thomas Braschler
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, CH-1022 Geneva, Switzerland; (F.B.); (A.B.); (A.B.)
| |
Collapse
|
21
|
Tuvshindorj U, Trouillet V, Vasilevich A, Koch B, Vermeulen S, Carlier A, Alexander MR, Giselbrecht S, Truckenmüller R, de Boer J. The Galapagos Chip Platform for High-Throughput Screening of Cell Adhesive Chemical Micropatterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105704. [PMID: 34985808 DOI: 10.1002/smll.202105704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/04/2021] [Indexed: 06/14/2023]
Abstract
In vivo cells reside in a complex extracellular matrix (ECM) that presents spatially distributed biochemical and -physical cues at the nano- to micrometer scales. Chemical micropatterning is successfully used to generate adhesive islands to control where and how cells attach and restore cues of the ECM in vitro. Although chemical micropatterning has become a powerful tool to study cell-material interactions, only a fraction of the possible micropattern designs was covered so far, leaving many other possible designs still unexplored. Here, a high-throughput screening platform called "Galapagos chip" is developed. It contains a library of 2176 distinct subcellular chemical patterns created using mathematical algorithms and a straightforward UV-induced two-step surface modification. This approach enables the immobilization of ligands in geometrically defined regions onto cell culture substrates. To validate the system, binary RGD/polyethylene glycol patterns are prepared on which human mesenchymal stem cells are cultured, and the authors observe how different patterns affect cell and organelle morphology. As proof of concept, the cells are stained for the mechanosensitive YAP protein, and, using a machine-learning algorithm, it is demonstrated that cell shape and YAP nuclear translocation correlate. It is concluded that the Galapagos chip is a versatile platform to screen geometrical aspects of cell-ECM interaction.
Collapse
Affiliation(s)
- Urandelger Tuvshindorj
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Vanessa Trouillet
- Institute for Applied Materials and Karlsruhe Nano Micro Facility, Karlsruhe Institute of Technology, 76344, Eggenstein-Leopoldshafen, Germany
| | - Aliaksei Vasilevich
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Britta Koch
- Advanced Materials and Healthcare Technologies Division, The School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Steven Vermeulen
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, The School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering and Institute, for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| |
Collapse
|
22
|
Michalak AL, Trieger GW, Trieger KA, Godula K. Stem Cell Microarrays for Assessing Growth Factor Signaling in Engineered Glycan Microenvironments. Adv Healthc Mater 2022; 11:e2101232. [PMID: 34541824 PMCID: PMC8854331 DOI: 10.1002/adhm.202101232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/19/2021] [Indexed: 02/03/2023]
Abstract
Extracellular glycans, such as glycosaminoglycans (GAGs), provide an essential regulatory component during the development and maintenance of tissues. GAGs, which harbor binding sites for a range of growth factors (GFs) and other morphogens, help establish gradients of these molecules in the extracellular matrix (ECM) and promote the formation of active signaling complexes when presented at the cell surface. As such, GAGs have been pursued as biologically active components for the development of biomaterials for cell-based regenerative therapies. However, their structural complexity and compositional heterogeneity make establishing structure-function relationships for this class of glycans difficult. Here, a stem cell array platform is described, in which chemically modified heparan sulfate (HS) GAG polysaccharides are conjugated to a gelatin matrix and introduced into a polyacrylamide hydrogel network. This array allowed for direct analysis of HS contributions to the signaling via the FGF2-dependent mitogen activated protein kinase (MAPK) pathway in mouse embryonic stem cells. With the recent emergence of powerful synthetic and recombinant technologies to produce well-defined GAG structures, a platform for analyzing both growth factor binding and signaling in response to the presence of these biomolecules will provide a powerful tool for integrating glycans into biomaterials to advance their biological properties and applications.
Collapse
Affiliation(s)
- Austen L. Michalak
- Deparment of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla CA 92093, USA
| | - Greg W. Trieger
- Deparment of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla CA 92093, USA
| | - Kelsey A. Trieger
- Deparment of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla CA 92093, USA
| | - Kamil Godula
- Deparment of Chemistry and Biochemistry, University of California San Diego, 9500 Gilman Drive, La Jolla CA 92093, USA,Glycobiology Research and Training Center, University of California San Diego, 9500 Gilman Drive, La Jolla CA 92093, USA
| |
Collapse
|
23
|
Mukundan S, Bell J, Teryek M, Hernandez C, Love AC, Parekkadan B, Chan LLY. Automated Assessment of Cancer Drug Efficacy On Breast Tumor Spheroids in Aggrewell™400 Plates Using Image Cytometry. J Fluoresc 2022; 32:521-531. [PMID: 34989923 DOI: 10.1007/s10895-021-02881-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/27/2021] [Indexed: 12/21/2022]
Abstract
Tumor spheroid models have proven useful in the study of cancer cell responses to chemotherapeutic compounds by more closely mimicking the 3-dimensional nature of tumors in situ. Their advantages are often offset, however, by protocols that are long, complicated, and expensive. Efforts continue for the development of high-throughput assays that combine the advantages of 3D models with the convenience and simplicity of traditional 2D monolayer methods. Herein, we describe the development of a breast cancer spheroid image cytometry assay using T47D cells in Aggrewell™400 spheroid plates. Using the Celigo® automated imaging system, we developed a method to image and individually track thousands of spheroids within the Aggrewell™400 microwell plate over time. We demonstrate the use of calcein AM and propidium iodide staining to study the effects of known anti-cancer drugs Doxorubicin, Everolimus, Gemcitabine, Metformin, Paclitaxel and Tamoxifen. We use the image cytometry results to quantify the fluorescence of calcein AM and PI as well as spheroid size in a dose dependent manner for each of the drugs. We observe a dose-dependent reduction in spheroid size and find that it correlates well with the viability obtained from the CellTiter96® endpoint assay. The image cytometry method we demonstrate is a convenient and high-throughput drug-response assay for breast cancer spheroids under 400 μm in diameter, and may lay a foundation for investigating other three-dimensional spheroids, organoids, and tissue samples.
Collapse
Affiliation(s)
- Shilpaa Mukundan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jordan Bell
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC, Lawrence, MA, 01843, USA
| | - Matthew Teryek
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Charles Hernandez
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC, Lawrence, MA, 01843, USA
| | - Andrea C Love
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC, Lawrence, MA, 01843, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.,Department of Medicine, Rutgers Biomedical Health Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Leo Li-Ying Chan
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC, Lawrence, MA, 01843, USA.
| |
Collapse
|
24
|
Soni B, Singh S. Synthetic Perturbations in IL6 Biological Circuit Induces Dynamical Cellular Response. Molecules 2021; 27:molecules27010124. [PMID: 35011356 PMCID: PMC8746995 DOI: 10.3390/molecules27010124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022] Open
Abstract
Macrophage phenotype plays a crucial role in the pathogenesis of Leishmanial infection. Pro-inflammatory cytokines signals through the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway that functions in parasite killing. Suppression of cytokine signaling (SOCS) is a well-known negative feedback regulator of the JAK/STAT pathway. However, change in the expression levels of SOCSs in correlation with the establishment of infection is not well understood. IL6 is a pleotropic cytokine that induces SOCS1 and SOCS3 expression through JAK-STAT signaling. Mathematical modeling of the TLR2 and IL6 signaling pathway has established the immune axis of SOCS1 and SOCS3 functioning in macrophage polarization during the early stage of Leishmania major infection. The ratio has been quantified both in silico and in vitro as 3:2 which is required to establish infection during the early stage. Furthermore, phosphorylated STAT1 and STAT3 have been established as an immunological cross talk between TLR2 and IL6 signaling pathways. Using synthetic biology approaches, peptide based immuno-regulatory circuits have been designed to target the activity of SOCS1 which can restore pro-inflammatory cytokine expression during infection. In a nutshell, we explored the potential of synthetic biology to address and rewire the immune response from Th2 to Th1 type during the early stage of leishmanial infection governed by SOCS1/SOCS3 immune axis.
Collapse
Affiliation(s)
| | - Shailza Singh
- Correspondence: or ; Tel.: +91-20-2570-8296 (ext. 95); Fax: +91-20-2569-2259
| |
Collapse
|
25
|
Liu H, Usprech JF, Parameshwar PK, Sun Y, Simmons CA. Combinatorial screen of dynamic mechanical stimuli for predictive control of MSC mechano-responsiveness. SCIENCE ADVANCES 2021; 7:7/19/eabe7204. [PMID: 33962940 PMCID: PMC8104874 DOI: 10.1126/sciadv.abe7204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/19/2021] [Indexed: 05/05/2023]
Abstract
Mechanobiological-based control of mesenchymal stromal cells (MSCs) to facilitate engineering and regeneration of load-bearing tissues requires systematic investigations of specific dynamic mechanical stimulation protocols. Using deformable membrane microdevice arrays paired with combinatorial experimental design and modeling, we probed the individual and integrative effects of mechanical stimulation parameters (strain magnitude, rate at which strain is changed, and duty period) on myofibrogenesis and matrix production of MSCs in three-dimensional hydrogels. These functions were found to be dominantly influenced by a previously unidentified, higher-order interactive effect between strain magnitude and duty period. Empirical models based on our combinatorial cue-response data predicted an optimal loading regime in which strain magnitude and duty period were increased synchronously over time, which was validated to most effectively promote MSC matrix production. These findings inform the design of loading regimes for MSC-based engineered tissues and validate a broadly applicable approach to probe multifactorial regulating effects of mechanobiological cues.
Collapse
Affiliation(s)
- Haijiao Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Jenna F Usprech
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Prabu Karthick Parameshwar
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
26
|
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: 94] [Impact Index Per Article: 23.5] [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
|
27
|
Balik K, Matulewicz K, Modrakowska P, Kozłowska J, Montane X, Tylkowski B, Bajek A. Advanced cell culture techniques for cancer research. PHYSICAL SCIENCES REVIEWS 2020. [DOI: 10.1515/psr-2019-0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The incessant increase number of cancer cases, motivates scientists to constantly develop and search for new therapies. Along with the dynamic development of anti-cancer drugs and therapies, we are witnessing huge progress in the world of science - the development of personalized medicine. An inseparable element is also a very strong trend in the development of new in vitro animal models for chemotherapeutic research. Cell cultures are commonly undertaken by research models before animal testing. They are the basis for the development of new diagnostic and cancer treatments. It should be emphasized that basic research is a strong foundation for any therapy introduced. This chapter provides an overview of the modern cell culture techniques that are currently developing, which allow the introduction of modern models that reflect the organs and physiological system. Currently available cell culture methods are a key aspect of studying these interactions, however, a method that eliminates the limitations of standard methods is still being sought.
Collapse
Affiliation(s)
- Karolina Balik
- Department of Tissue Engineering Chair of Urology , Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun , Bydgoszcz , Poland
| | - Karolina Matulewicz
- Department of Tissue Engineering Chair of Urology , Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun , Bydgoszcz , Poland
| | - Paulina Modrakowska
- Department of Tissue Engineering Chair of Urology , Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun , Bydgoszcz , Poland
| | - Jolanta Kozłowska
- Department of Biological Pest Control and Organic Agriculture , Institute of Plant Protection, National Research Institute , Poznan , Poland
| | - Xavier Montane
- Department of Chemical Engineering , Universitat Rovira i Virgili , Tarragona , Spain
| | - Bartosz Tylkowski
- Chemical Technologies Unit , Eurecat, Centre Tecnològic de Catalunya , Tarragona , Spain
| | - Anna Bajek
- Department of Tissue Engineering Chair of Urology , Ludwik Rydygier Collegium Medicum in Bydgoszcz Nicolaus Copernicus University in Torun , Bydgoszcz , Poland
| |
Collapse
|
28
|
Li Y, Wang S, Dong Y, Mu P, Yang Y, Liu X, Lin C, Huang Q. Effect of size and crystalline phase of TiO 2 nanotubes on cell behaviors: A high throughput study using gradient TiO 2 nanotubes. Bioact Mater 2020; 5:1062-1070. [PMID: 32695936 PMCID: PMC7363987 DOI: 10.1016/j.bioactmat.2020.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
The research of TiO2 nanotubes (TNTs) in the field of biomedicine has been increasingly active. However, given the diversity of the nanoscale dimension and controversial reports, our understanding of the structure-property relationships of TNTs is not yet complete. In this paper, gradient TNTs with a wide diameter range of 20-350 nm were achieved by bipolar electrochemistry and utilized for a thorough high-throughput study of the effect of nanotube dimension and crystalline phase on protein adsorption and cell behaviors. Results indicated that protein adsorption escalated with nanotube dimension whereas cell proliferation and differentiation are preferred on small diameter (<70 nm) nanotubes. Large diameter anatase nanotubes had higher adsorption of serum proteins than as-prepared ones. But only as-prepared small diameter nanotubes presented slightly higher cell proliferation than corresponding annealed nanotubes whereas there was no discernible difference between as-prepared and annealed nanotubes on cell differentiation for the entire gradient. Those findings replenish previous research about how cell responses to TNTs with a wide diameter range and provide scientific guidance for the optimal design of biomedical materials.
Collapse
Affiliation(s)
- Yanran Li
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Si Wang
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Yuanjun Dong
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Ping Mu
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Yun Yang
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| | - Xiangyang Liu
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
- Department of Physics, National University of Singapore, 2 Science Drive 3, 117542, Singapore
| | - Changjian Lin
- State Key Laboratory for Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Qiaoling Huang
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
29
|
Li Y, Liu H, Cui Y, Chen H, Cui X, Shao J, Su F, He X. miR-424-3p Contributes to the Malignant Progression and Chemoresistance of Gastric Cancer. Onco Targets Ther 2020; 13:12201-12211. [PMID: 33273826 PMCID: PMC7705957 DOI: 10.2147/ott.s280717] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/09/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common and lethal malignancies worldwide. Therefore, a better understanding of the mechanism of its malignant progression and chemoresistance will be helpful for the treatment of patients with GC. METHODS The gene expression profiles downloaded from GEO database and the TargetScan Human were used to identify the key regulation model based on miRNA by bioinformatics analyses. The regulation of miRNA to target was clarified by luciferase assay, qPCR, and Western blotting. Then, the in vitro and in vivo experiments were further conducted by overexpression or knockdown of miRNA and/or target to examine the regulation effects and clarify the mechanism. RESULTS In the present study, miR-424-3p was identified to be differentially expressed among normal gastric, GC, and chemoresistant GC tissues. Target analysis results indicated that ABCC2, a chemoresistance-related gene, was a regulated target of miR-424-3p. The in vitro and in vivo experiment results further demonstrated that miR-424-3p relied on ABCC2-induced chemoresistance to promote GC proliferation and metastasis. CONCLUSION Overall, this study revealed that miR-424-3p contributed to the malignant progression and chemoresistance of GC. Thus, miR-424-3p could be a potential target for the treatment of GC.
Collapse
Affiliation(s)
- Yongyuan Li
- Department of General Surgery, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin300052, People’s Republic of China
| | - Hongjie Liu
- Department of Radiology, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
| | - Yu Cui
- Department of General Surgery, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
| | - Hekai Chen
- Department of General Surgery, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
| | - Xuejun Cui
- Department of General Surgery, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
| | - Jianping Shao
- Department of General Surgery, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
| | - Feng Su
- Department of General Surgery, The Fifth Central Hospital, Tianjin300450, People’s Republic of China
| | - Xianghui He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin300052, People’s Republic of China
| |
Collapse
|
30
|
Muckom RJ, Sampayo RG, Johnson HJ, Schaffer DV. Advanced Materials to Enhance Central Nervous System Tissue Modeling and Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002931. [PMID: 33510596 PMCID: PMC7840150 DOI: 10.1002/adfm.202002931] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 05/04/2023]
Abstract
The progressively deeper understanding of mechanisms underlying stem cell fate decisions has enabled parallel advances in basic biology-such as the generation of organoid models that can further one's basic understanding of human development and disease-and in clinical translation-including stem cell based therapies to treat human disease. Both of these applications rely on tight control of the stem cell microenvironment to properly modulate cell fate, and materials that can be engineered to interface with cells in a controlled and tunable manner have therefore emerged as valuable tools for guiding stem cell growth and differentiation. With a focus on the central nervous system (CNS), a broad range of material solutions that have been engineered to overcome various hurdles in constructing advanced organoid models and developing effective stem cell therapeutics is reviewed. Finally, regulatory aspects of combined material-cell approaches for CNS therapies are considered.
Collapse
Affiliation(s)
- Riya J Muckom
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Rocío G Sampayo
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| | - Hunter J Johnson
- Department of Bioengineering, UC Berkeley, Berkeley, CA 94704, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, UC Berkeley, Berkeley, CA 94704, USA
| |
Collapse
|
31
|
Hall MS, Decker JT, Shea LD. Towards systems tissue engineering: Elucidating the dynamics, spatial coordination, and individual cells driving emergent behaviors. Biomaterials 2020; 255:120189. [PMID: 32569865 PMCID: PMC7396312 DOI: 10.1016/j.biomaterials.2020.120189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/20/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022]
Abstract
Biomaterial systems have enabled the in vitro production of complex, emergent tissue behaviors that were not possible with conventional two-dimensional culture systems, allowing for analysis of both normal development and disease processes. We propose that the path towards developing the design parameters for biomaterial systems lies with identifying the molecular drivers of emergent behavior through leveraging technological advances in systems biology, including single cell omics, genetic engineering, and high content imaging. This growing research opportunity at the intersection of the fields of tissue engineering and systems biology - systems tissue engineering - can uniquely interrogate the mechanisms by which complex tissue behaviors emerge with the potential to capture the contribution of i) dynamic regulation of tissue development and dysregulation, ii) single cell heterogeneity and the function of rare cell types, and iii) the spatial distribution and structure of individual cells and cell types within a tissue. By leveraging advances in both biological and materials data science, systems tissue engineering can facilitate the identification of biomaterial design parameters that will accelerate basic science discovery and translation.
Collapse
Affiliation(s)
- Matthew S Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
Amin Yavari S, Castenmiller SM, van Strijp JAG, Croes M. Combating Implant Infections: Shifting Focus from Bacteria to Host. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002962. [PMID: 32914481 DOI: 10.1002/adma.202002962] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Indexed: 05/06/2023]
Abstract
The widespread use of biomaterials to support or replace body parts is increasingly threatened by the risk of implant-associated infections. In the quest for finding novel anti-infective biomaterials, there generally has been a one-sided focus on biomaterials with direct antibacterial properties, which leads to excessive use of antibacterial agents, compromised host responses, and unpredictable effectiveness in vivo. This review sheds light on how host immunomodulation, rather than only targeting bacteria, can endow biomaterials with improved anti-infective properties. How antibacterial surface treatments are at risk to be undermined by biomaterial features that dysregulate the protection normally provided by critical immune cell subsets, namely, neutrophils and macrophages, is discussed. Accordingly, how the precise modification of biomaterial surface biophysical cues, or the incorporation of immunomodulatory drug delivery systems, can render biomaterials with the necessary immune-compatible and immune-protective properties to potentiate the host defense mechanisms is reviewed. Within this context, the protective role of host defense peptides, metallic particles, quorum sensing inhibitors, and therapeutic adjuvants is discussed. The highlighted immunomodulatory strategies may lay a foundation to develop anti-infective biomaterials, while mitigating the increasing threat of antibacterial drug resistance.
Collapse
Affiliation(s)
- Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Suzanne M Castenmiller
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| |
Collapse
|
33
|
Sarwat M, Surrao DC, Huettner N, St John JA, Dargaville TR, Forget A. Going beyond RGD: screening of a cell-adhesion peptide library in 3D cell culture. Biomed Mater 2020; 15:055033. [DOI: 10.1088/1748-605x/ab9d6e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
Suvarnapathaki S, Wu X, Lantigua D, Nguyen MA, Camci-Unal G. Hydroxyapatite-Incorporated Composite Gels Improve Mechanical Properties and Bioactivity of Bone Scaffolds. Macromol Biosci 2020; 20:e2000176. [PMID: 32755044 DOI: 10.1002/mabi.202000176] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Reinforcing polymeric scaffolds with micro/nanoparticles improve their mechanical properties and render them bioactive. In this study, hydroxyapatite (HA) is incorporated into 5% (w/v) gelatin methacrylate (GelMA) hydrogels at 1, 5, and 20 mg mL-1 concentrations. The material properties of these composite gels are characterized through swelling, degradation, and compression tests. Using 3D cell encapsulation, the cytocompatibility and osteogenic differentiation of preosteoblasts are evaluated to assess the biological properties of the composite scaffolds. The in vitro assays demonstrate increasing cell proliferation and metabolic activity over the course of 14 d in culture. Furthermore, the scaffolds support osteogenic differentiation of the microencapsulated preosteoblasts. For the in vivo study, the composite scaffolds are subcutaneously implanted in rats for 14 d. The histological staining of the explanted in vivo samples exhibits the functional advantages of the scaffold's biocompatibility, biodegradability, and integration into the existing host tissue. This work demonstrates the enhanced mechanical and biological performance of HA-gelatin composite hydrogels for bone tissue engineering applications.
Collapse
Affiliation(s)
- Sanika Suvarnapathaki
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA.,Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA
| | - Xinchen Wu
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA.,Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA
| | - Darlin Lantigua
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA.,Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA
| | - Michelle A Nguyen
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts Lowell, One University Avenue, Lowell, MA, 01854, USA.,Department of Surgery, University of Massachusetts Medical School, 55 Lake Avenue, Worcester, MA, 01655, USA
| |
Collapse
|
35
|
Hur W, Son SE, Seong GH. Electrochemical live cell patterning. Electrochem commun 2020. [DOI: 10.1016/j.elecom.2020.106778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
36
|
Sakthivel K, Kumar H, Mohamed MGA, Talebjedi B, Shim J, Najjaran H, Hoorfar M, Kim K. High Throughput Screening of Cell Mechanical Response Using a Stretchable 3D Cellular Microarray Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000941. [PMID: 32588966 DOI: 10.1002/smll.202000941] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/10/2020] [Indexed: 06/11/2023]
Abstract
Cells in vivo are constantly subjected to multiple microenvironmental mechanical stimuli that regulate cell function. Although 2D cell responses to the mechanical stimulation have been established, these methods lack relevance as physiological cell microenvironments are in 3D. Moreover, the existing platforms developed for studying the cell responses to mechanical cues in 3D either offer low-throughput, involve complex fabrication, or do not allow combinatorial analysis of multiple cues. Considering this, a stretchable high-throughput (HT) 3D cell microarray platform is presented that can apply dynamic mechanical strain to cells encapsulated in arrayed 3D microgels. The platform uses inkjet-bioprinting technique for printing cell-laden gelatin methacrylate (GelMA) microgel array on an elastic composite substrate that is periodically stretched. The developed platform is highly biocompatible and transfers the applied strain from the stretched substrate to the cells. The HT analysis is conducted to analyze cell mechano-responses throughout the printed microgel array. Also, the combinatorial analysis of distinct cell behaviors is conducted for different GelMA microenvironmental stiffnesses in addition to the dynamic stretch. Considering its throughput and flexibility, the developed platform can readily be scaled up to introduce a wide range of microenvironmental cues and to screen the cell responses in a HT way.
Collapse
Affiliation(s)
- Kabilan Sakthivel
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Hitendra Kumar
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Mohamed G A Mohamed
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Bahram Talebjedi
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Justin Shim
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Homayoun Najjaran
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Mina Hoorfar
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Keekyoung Kim
- School of Engineering, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
- Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB, T2N 1N4, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, T2N 1N4, Canada
| |
Collapse
|
37
|
Rima XY, Walters N, Nguyen LTH, Reátegui E. Surface engineering within a microchannel for hydrodynamic and self-assembled cell patterning. BIOMICROFLUIDICS 2020; 14:014104. [PMID: 31933714 PMCID: PMC6941948 DOI: 10.1063/1.5126608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/18/2019] [Indexed: 05/27/2023]
Abstract
The applications of cell patterning are widespread due to the high-throughput testing and different resolutions offered by these platforms. Cell patterning has aided in deconvoluting in vivo experiments to better characterize cellular mechanisms and increase therapeutic output. Here, we present a technique for engineering an artificial surface via surface chemistry to form large-scale arrays of cells within a microchannel by employing microstamping. By changing the approach in surface chemistry, H1568 cells were patterned hydrodynamically using immunoaffinity, and neutrophils were patterned through self-assembly via chemotaxis. The high patterning efficiencies (93% for hydrodynamic patterning and 68% for self-assembled patterning) and the lack of secondary adhesion demonstrate the reproducibility of the platform. The interaction between H1568 and neutrophils was visualized and quantified to determine the capability of the platform to encourage cell-cell interaction. With the introduction of H1568 cells into the self-assembled patterning platform, a significant hindrance in the neutrophils' ability to swarm was observed, indicating the important roles of inflammatory mediators within the nonsmall cell lung cancer tumor microenvironment.
Collapse
Affiliation(s)
- Xilal Y. Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Nicole Walters
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Luong T. H. Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
38
|
Zeng W, Xu B, Du G, Chen J, Zhou J. Integrating enzyme evolution and high-throughput screening for efficient biosynthesis of l-DOPA. ACTA ACUST UNITED AC 2019; 46:1631-1641. [DOI: 10.1007/s10295-019-02237-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/11/2019] [Indexed: 02/03/2023]
Abstract
Abstract
l-DOPA is a key pharmaceutical agent for treating Parkinson’s, and market demand has exploded due to the aging population. There are several challenges associated with the chemical synthesis of l-DOPA, including complicated operation, harsh conditions, and serious pollution. A biocatalysis route for l-DOPA production is promising, especially via a route catalyzed by tyrosine phenol lyase (TPL). In this study, using TPL derived from Erwinia herbicola (Eh-TPL), a mutant Eh-TPL was obtained by integrating enzyme evolution and high-throughput screening methods. l-DOPA production using recombinant Escherichia coli BL21 (DE3) cells harbouring mutant Eh-TPL was enhanced by 36.5% in shake flasks, and the temperature range and alkali resistance of the Eh-TPL mutant were promoted. Sequence analysis revealed two mutated amino acids in the mutant (S20C and N161S), which reduced the length of a hydrogen bond and generated new hydrogen bonds. Using a fed-batch mode for whole-cell catalysis in a 5 L bioreactor, the titre of l-DOPA reached 69.1 g L−1 with high productivity of 11.52 g L−1 h−1, demonstrating the great potential of Eh-TPL variants for industrial production of l-DOPA.
Collapse
Affiliation(s)
- Weizhu Zeng
- grid.258151.a 0000 0001 0708 1323 Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 National Engineering Laboratory for Cereal Fermentation Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
| | - Bingbing Xu
- grid.258151.a 0000 0001 0708 1323 Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 National Engineering Laboratory for Cereal Fermentation Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 Jiangsu Provisional Research Center for Bioactive Product Processing Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
| | - Guocheng Du
- grid.258151.a 0000 0001 0708 1323 Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology Jiangnan University 214122 Wuxi China
| | - Jian Chen
- grid.258151.a 0000 0001 0708 1323 Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 National Engineering Laboratory for Cereal Fermentation Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 Jiangsu Provisional Research Center for Bioactive Product Processing Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
| | - Jingwen Zhou
- grid.258151.a 0000 0001 0708 1323 Key Laboratory of Industrial Biotechnology, Ministry of Education and School of Biotechnology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 National Engineering Laboratory for Cereal Fermentation Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
- grid.258151.a 0000 0001 0708 1323 Jiangsu Provisional Research Center for Bioactive Product Processing Technology Jiangnan University 1800 Lihu Road 214122 Wuxi Jiangsu China
| |
Collapse
|
39
|
Zhou M, Wu Z, Zhao Y, Yang Q, Ling W, Li Y, Xu H, Wang C, Huang X. Droplets as Carriers for Flexible Electronic Devices. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901862. [PMID: 31871863 PMCID: PMC6918117 DOI: 10.1002/advs.201901862] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/16/2019] [Indexed: 05/30/2023]
Abstract
Coupling soft bodies and dynamic motions with multifunctional flexible electronics is challenging, but is essential in satisfying the urgent and soaring demands of fully soft and comprehensive robotic systems that can perform tasks in spite of rigorous spatial constraints. Here, the mobility and adaptability of liquid droplets with the functionality of flexible electronics, and techniques to use droplets as carriers for flexible devices are combined. The resulting active droplets (ADs) with volumes ranging from 150 to 600 µL can conduct programmable functions, such as sensing, actuation, and energy harvesting defined by the carried flexible devices and move under the excitation of gravitational force or magnetic force. They work in both dry and wet environments, and adapt to the surrounding environment through reversible shape shifting. These ADs can achieve controllable motions at a maximum velocity of 226 cm min-1 on a dry surface and 32 cm min-1 in a liquid environment. The conceptual system may eventually lead to individually addressable ADs that offer sophisticated functions for high-throughput molecule analysis, drug assessment, chemical synthesis, and information collection.
Collapse
Affiliation(s)
- Mingxing Zhou
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Ziyue Wu
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Yicong Zhao
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Qing Yang
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Wei Ling
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Ya Li
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Hang Xu
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| | - Cheng Wang
- Department of Mechanical EngineeringMissouri University of Science and Technology400 West 13th StreetRollaMO65401USA
| | - Xian Huang
- Department of Biomedical EngineeringTianjin University92 Weijin RoadTianjin300072P. R. China
| |
Collapse
|
40
|
Nongenetic optical modulation of neural stem cell proliferation and neuronal/glial differentiation. Biomaterials 2019; 225:119539. [PMID: 31622821 DOI: 10.1016/j.biomaterials.2019.119539] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/01/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
Abstract
Photostimulation has been widely used in neuromodulation. However, existing optogenetics techniques require genetic alternation of the targeted cell or tissue. Here, we report that neural stem cells (NSCs) constitutionally express blue/red light-sensitive photoreceptors. The proliferation and regulation of NSCs to neuronal or glial cells are wavelength-specific. Our results showed a 4.3-fold increase in proliferation and 2.7-fold increase in astrocyte differentiation for cells under low-power blue monochromatic light exposure (455 nm, 300 μW/cm2). The melanopsin (Opn4)/transient receptor potential channel 6 (TRPC6) non-visual opsin serves as a key photoreceptor response to blue light irradiation. Two-dimensional gel electrophoresis coupled with mass spectrometry further highlighted the Jun activation domain-binding protein 1 (Jab1) as a novel and specific modulator in phototransduction pathways induced by blue light exposure. Quiescent adult NSCs reside in specific regions of the mammalian brain. Therefore, we showed that melanopsin/TRPC6 expressed in these regions and blue light stimulation through optical fibers could directly stimulate the NSCs in vivo. Upconversion nanoparticles (UCNPs) converted deep-penetrating near-infrared (NIR) light into specific wavelengths of visible light. Accordingly, we demonstrated that UCNP-mediated NIR light could be used to modulate in vivo NSC differentiation in a less invasive manner. In the future, this light-triggered system of NSCs will enable nongenetic and noninvasive neuromodulation with therapeutic potential for central nervous system diseases.
Collapse
|
41
|
Park K, Lee Y, Seo J. Recent Advances in High-throughput Platforms with Engineered Biomaterial Microarrays for Screening of Cell and Tissue Behavior. Curr Pharm Des 2019; 24:5458-5470. [DOI: 10.2174/1381612825666190207093438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 02/06/2023]
Abstract
In the last decades, bioengineers have developed myriad biomaterials for regenerative medicine. Development of screening techniques is essential for understanding complex behavior of cells in the biological microenvironments. Conventional approaches to the screening of cellular behavior in vitro have limitations in terms of accuracy, reusability, labor-intensive screening, and versatility. Thus, drug screening and toxicology test through in vitro screening platforms have been underwhelming. Recent advances in the high-throughput screening platforms somewhat overcome the limitations of in vitro screening platforms via repopulating human tissues’ biophysical and biomchemical microenvironments with the ability to continuous monitoring of miniaturized human tissue behavior. Herein, we review current trends in the screening platform in which a high-throughput system composed of engineered microarray devices is developed to investigate cell-biomaterial interaction. Furthermore, diverse methods to achieve continuous monitoring of cell behavior via developments of biosensor integrated high-throughput platforms, and future perspectives on high-throughput screening will be provided.
Collapse
Affiliation(s)
- Kijun Park
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Yeontaek Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Jungmok Seo
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| |
Collapse
|
42
|
Bandaru P, Chu D, Sun W, Lasli S, Zhao C, Hou S, Zhang S, Ni J, Cefaloni G, Ahadian S, Dokmeci MR, Sengupta S, Lee J, Khademhosseini A. A Microfabricated Sandwiching Assay for Nanoliter and High-Throughput Biomarker Screening. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900300. [PMID: 30884183 DOI: 10.1002/smll.201900300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/14/2019] [Indexed: 05/03/2023]
Abstract
Cells secrete substances that are essential to the understanding of numerous immunological phenomena and are extensively used in clinical diagnoses. Countless techniques for screening of biomarker secretion in living cells have generated valuable information on cell function and physiology, but low volume and real-time analysis is a bottleneck for a range of approaches. Here, a simple, highly sensitive assay using a high-throughput micropillar and microwell array chip (MIMIC) platform is presented for monitoring of biomarkers secreted by cancer cells. The sensing element is a micropillar array that uses the enzyme-linked immunosorbent assay (ELISA) mechanism to detect captured biomolecules. When integrated with a microwell array where few cells are localized, interleukin 8 (IL-8) secretion can be monitored with nanoliter volume using multiple micropillar arrays. The trend of cell secretions measured using MIMICs matches the results from conventional ELISA well while it requires orders of magnitude less cells and volumes. Moreover, the proposed MIMIC is examined to be used as a drug screening platform by delivering drugs using micropillar arrays in combination with a microfluidic system and then detecting biomolecules from cells as exposed to drugs.
Collapse
Affiliation(s)
- Praveen Bandaru
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Dafeng Chu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Wujin Sun
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Soufian Lasli
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Chuanzhen Zhao
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Shuang Hou
- Department of Molecular and Medical Pharmacology, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Shiming Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Jiahua Ni
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Giorgia Cefaloni
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Samad Ahadian
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Mehmet Remzi Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Shiladitya Sengupta
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Junmin Lee
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Ali Khademhosseini
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
43
|
Li Y, Dong Y, Yang Y, Yu P, Zhang Y, Hu J, Li T, Zhang X, Liu X, Xu Q, Huang Q, Lin C. Rational Design of Silver Gradient for Studying Size Effect of Silver Nanoparticles on Contact Killing. ACS Biomater Sci Eng 2019; 5:425-431. [PMID: 33405808 DOI: 10.1021/acsbiomaterials.8b01282] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The cellular mechanism underlying bacteria responses to silver nanoparticles (AgNPs) has not been fully elucidated. Especially, it is difficult to distinguish the contact killing from release killing as Ag+ releases from AgNPs. In this paper, AgNPs gradient was designed for evaluating the size effect of AgNPs on contact killing. A size gradient of AgNPs (5-45 nm) was achieved on TiO2 nanotubes (TNTs) by rational design of bipolar electrochemical reaction, including applied voltage, electrolyte concentration, and sample size. High-throughput investigation of cellular responses showed that the smallest AgNPs were the most efficient in suppressing bacteria whereas the largest AgNPs were more favorable for MC3T3-E1 cell adhesion and proliferation. As Ag+ concentration was the same for the entire gradient, the difference in cellular responses was dominated by the contact effect (rather than difference in released Ag+) which was tuned by AgNPs size. This method offers new prospect for efficient evaluation of the contact effect of nanoparticles, such as Ag, Au, and Cu.
Collapse
Affiliation(s)
- Yanran Li
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | - Yuanjun Dong
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | - Yun Yang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | - Ping Yu
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | | | | | - Tang Li
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Xiangyang Liu
- Department of Physics, National University of Singapore, 2 Science Drive 3, Singapore, 117542, Singapore
| | - Qingchi Xu
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | - Qiaoling Huang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
| | | |
Collapse
|
44
|
Qi X, Wei W, Shen J, Dong W. Salecan polysaccharide-based hydrogels and their applications: a review. J Mater Chem B 2019; 7:2577-2587. [PMID: 32254990 DOI: 10.1039/c8tb03312a] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This review systematically summarizes for the first time the recent progress on hydrogels containing salecan polysaccharides.
Collapse
Affiliation(s)
- Xiaoliang Qi
- School of Ophthalmology & Optometry
- Eye Hospital
- Wenzhou Medical University
- Wenzhou
- China
| | - Wei Wei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine
- and Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province
- First Affiliated Hospital
- Zhejiang University School of Medicine
- Hangzhou
| | - Jianliang Shen
- School of Ophthalmology & Optometry
- Eye Hospital
- Wenzhou Medical University
- Wenzhou
- China
| | - Wei Dong
- Center for Molecular Metabolism
- Nanjing University of Science & Technology
- Nanjing 210094
- China
| |
Collapse
|
45
|
Campbell JM, Balhoff JB, Landwehr GM, Rahman SM, Vaithiyanathan M, Melvin AT. Microfluidic and Paper-Based Devices for Disease Detection and Diagnostic Research. Int J Mol Sci 2018; 19:E2731. [PMID: 30213089 PMCID: PMC6164778 DOI: 10.3390/ijms19092731] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Recent developments in microfluidic devices, nanoparticle chemistry, fluorescent microscopy, and biochemical techniques such as genetic identification and antibody capture have provided easier and more sensitive platforms for detecting and diagnosing diseases as well as providing new fundamental insight into disease progression. These advancements have led to the development of new technology and assays capable of easy and early detection of pathogenicity as well as the enhancement of the drug discovery and development pipeline. While some studies have focused on treatment, many of these technologies have found initial success in laboratories as a precursor for clinical applications. This review highlights the current and future progress of microfluidic techniques geared toward the timely and inexpensive diagnosis of disease including technologies aimed at high-throughput single cell analysis for drug development. It also summarizes novel microfluidic approaches to characterize fundamental cellular behavior and heterogeneity.
Collapse
Affiliation(s)
- Joshua M Campbell
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Joseph B Balhoff
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Grant M Landwehr
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Sharif M Rahman
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| | | | - Adam T Melvin
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
46
|
Zhou J, Du X, Chen X, Xu B. Adaptive Multifunctional Supramolecular Assemblies of Glycopeptides Rapidly Enable Morphogenesis. Biochemistry 2018; 57:4867-4879. [PMID: 30001488 PMCID: PMC6092213 DOI: 10.1021/acs.biochem.8b00125] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the well-established biophysical principle of adhesion-guided in vitro morphogenesis, there are few single synthetic molecular species that can rapidly enable morphogenesis (e.g., a cell monolayer to cell spheroids) in a cell culture because adhesion inherently involves many signals. Here we show the use of adaptive multifunctional supramolecular assemblies of glycopeptides, consisting of cell adhesion sequence and saccharide, to induce cell spheroids rapidly from a monolayer of cells. Having a general architecture of N-terminal capping, glycosylation, and an integrin-binding sequence, the glycopeptides self-assemble to form a dynamic continuum of nanostructures (i.e., from nanoparticles to nanofibers) to affect the interactions of integrins, E-selectin, and cadherins with their natural ligands and to act adaptively according to the cellular environment. Such adaptive (i.e., context-dependent) interactions weaken cell-substratum adhesion and enhance intercellular interactions, which rapidly and transiently induce cell spheroids. This work illustrates the use of supramolecular assemblies of simple glycopeptides to modulate biophysical conditions for regulating cell functions, which is a new approach for developing biomaterials.
Collapse
Affiliation(s)
| | | | - Xiaoyi Chen
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| |
Collapse
|
47
|
Machillot P, Quintal C, Dalonneau F, Hermant L, Monnot P, Matthews K, Fitzpatrick V, Liu J, Pignot-Paintrand I, Picart C. Automated Buildup of Biomimetic Films in Cell Culture Microplates for High-Throughput Screening of Cellular Behaviors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801097. [PMID: 29786885 PMCID: PMC6701983 DOI: 10.1002/adma.201801097] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/20/2018] [Indexed: 05/22/2023]
Abstract
An automatic method is established for layer-by-layer (LbL) assembly of biomimetic coatings in cell culture microplates using a commercial liquid-handling robot. Highly homogeneous thin films are formed at the bottom of each microwell. The LbL film-coated microplates are compatible with common cellular assays, using microplate readers and automated microscopes. Cellular adhesion is screened on crosslinked and peptide-functionalized LbL films and stem cell differentiation in response to increasing doses of bone morphogenetic proteins (2, 4, 7, 9). This method paves the way for future applications of LbL films in cell-based assays for regenerative medicine and high-throughput drug screening.
Collapse
Affiliation(s)
- Paul Machillot
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Catarina Quintal
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Fabien Dalonneau
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Loic Hermant
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Pauline Monnot
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Kelsey Matthews
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Vincent Fitzpatrick
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Jie Liu
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Isabelle Pignot-Paintrand
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
| | - Catherine Picart
- Grenoble Institute of Technology, Univ. Grenoble Alpes, 38000 Grenoble, France
- CNRS, LMGP, 3 parvis Louis Néel, 38031 Grenoble Cedex 01, France
- Corresponding author:
| |
Collapse
|
48
|
Brooks EA, Jansen LE, Gencoglu MF, Yurkevicz AM, Peyton SR. Complementary, Semiautomated Methods for Creating Multidimensional PEG-Based Biomaterials. ACS Biomater Sci Eng 2018; 4:707-718. [PMID: 33418758 DOI: 10.1021/acsbiomaterials.7b00737] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tunable biomaterials that mimic selected features of the extracellular matrix (ECM) such as its stiffness, protein composition, and dimensionality are increasingly popular for studying how cells sense and respond to ECM cues. In the field, there exists a significant trade-off for how complex and how well these biomaterials represent the in vivo microenvironment versus how easy they are to make and how adaptable they are to automated fabrication techniques. To address this need to integrate more complex biomaterials design with high-throughput screening approaches, we present several methods to fabricate synthetic biomaterials in 96-well plates and demonstrate that they can be adapted to semiautomated liquid handling robotics. These platforms include (1) glass bottom plates with covalently attached ECM proteins and (2) hydrogels with tunable stiffness and protein composition with either cells seeded on the surface or (3) laden within the three-dimensional hydrogel matrix. This study includes proof-of-concept results demonstrating control over breast cancer cell line phenotypes via these ECM cues in a semiautomated fashion. We foresee the use of these methods as a mechanism to bridge the gap between high-throughput cell-matrix screening and engineered ECM-mimicking biomaterials.
Collapse
Affiliation(s)
- Elizabeth A Brooks
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Lauren E Jansen
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Maria F Gencoglu
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Annali M Yurkevicz
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, N540 Life Sciences Laboratories, 240 Thatcher Road, Amherst, Massachusetts 01003-9364, United States
| |
Collapse
|
49
|
Smith Callahan LA. Gradient Material Strategies for Hydrogel Optimization in Tissue Engineering Applications. High Throughput 2018; 7:E1. [PMID: 29485612 PMCID: PMC5876527 DOI: 10.3390/ht7010001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022] Open
Abstract
Although a number of combinatorial/high-throughput approaches have been developed for biomaterial hydrogel optimization, a gradient sample approach is particularly well suited to identify hydrogel property thresholds that alter cellular behavior in response to interacting with the hydrogel due to reduced variation in material preparation and the ability to screen biological response over a range instead of discrete samples each containing only one condition. This review highlights recent work on cell-hydrogel interactions using a gradient material sample approach. Fabrication strategies for composition, material and mechanical property, and bioactive signaling gradient hydrogels that can be used to examine cell-hydrogel interactions will be discussed. The effects of gradients in hydrogel samples on cellular adhesion, migration, proliferation, and differentiation will then be examined, providing an assessment of the current state of the field and the potential of wider use of the gradient sample approach to accelerate our understanding of matrices on cellular behavior.
Collapse
Affiliation(s)
- Laura A Smith Callahan
- The Vivian L. Smith Department of Neurosurgery, Center for Stem Cell & Regenerative Medicine, and Department of Nanomedicine and Biomedical Engineering, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|