1
|
Ortiz-Cárdenas JE, Zatorski JM, Arneja A, Montalbine AN, Munson JM, Luckey CJ, Pompano RR. Towards spatially-organized organs-on-chip: Photopatterning cell-laden thiol-ene and methacryloyl hydrogels in a microfluidic device. ORGANS-ON-A-CHIP 2022; 4:100018. [PMID: 35535262 PMCID: PMC9078144 DOI: 10.1016/j.ooc.2022.100018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Micropatterning techniques for 3D cell cultures enable the recreation of tissue-level structures, but the combination of patterned hydrogels with organs-on-chip to generate organized 3D cultures under microfluidic perfusion remains challenging. To address this technological gap, we developed a user-friendly in-situ micropatterning protocol that integrates photolithography of crosslinkable, cell-laden hydrogels with a simple microfluidic housing, and tested the impact of crosslinking chemistry on stability and spatial resolution. Working with gelatin functionalized with photo-crosslinkable moieties, we found that inclusion of cells at high densities (≥ 107/mL) did not impede thiol-norbornene gelation, but decreased the storage moduli of methacryloyl hydrogels. Hydrogel composition and light dose were selected to match the storage moduli of soft tissues. To generate the desired pattern on-chip, the cell-laden precursor solution was flowed into a microfluidic chamber and exposed to 405 nm light through a photomask. The on-chip 3D cultures were self-standing and the designs were interchangeable by simply swapping out the photomask. Thiol-ene hydrogels yielded highly accurate feature sizes from 100 - 900 μm in diameter, whereas methacryloyl hydrogels yielded slightly enlarged features. Furthermore, only thiol-ene hydrogels were mechanically stable under perfusion overnight. Repeated patterning readily generated multi-region cultures, either separately or adjacent, including non-linear boundaries that are challenging to obtain on-chip. As a proof-of-principle, primary human T cells were patterned on-chip with high regional specificity. Viability remained high (> 85%) after 12-hr culture with constant perfusion. We envision that this technology will enable researchers to pattern 3D co-cultures to mimic organ-like structures that were previously difficult to obtain.
Collapse
Affiliation(s)
| | - Jonathan M. Zatorski
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, VA, USA 22904
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
| | - Jennifer M. Munson
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, VA, USA 22904
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
- Department of Chemistry, Carter Immunology Center, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
| |
Collapse
|
2
|
Additive Soft Matter Design by UV-Induced Polymer Hydrogel Inter-Crosslinking. Gels 2022; 8:gels8020117. [PMID: 35200499 PMCID: PMC8871859 DOI: 10.3390/gels8020117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/02/2022] Open
Abstract
In recent years, stimuli-responsive hydrogels have gained tremendous interest in designing complex smart 4D materials for applications ranging from biomedicine to soft electronics that can change their properties on demand over time. However, at present, a hydrogel’s response is often induced by merely a single stimulus, restricting its broader applicability. The controlled hierarchical assembly of various hydrogel building blocks, each with a tailored set of mechanical and physicochemical properties as well as programmed stimulus response, may potentially enable the design and fabrication of multi-responsive polymer parts that process complex operations, like signal routing dependent on different stimuli. Since inter-connection stability of such building blocks directly accompanies the transmission of information across building blocks and is as important as the building property itself to create complex 4D materials, we provide a study on the utility of an inter-crosslinking mechanism based on UV-induced 2,3-dimethylmaleimide (DMMI) dimerization to inter-connect acrylamide-based and N-isopropylacrylamide-based millimeter-sized cubic building blocks, respectively. The resulting dual-crosslinked assemblies are freestanding and stable against contraction–expansion cycles in solution. In addition, the approach is also applicable for connecting microfluidically fabricated, micrometer-sized hydrogel spheres, with the resulting assemblies being processable and mechanical stable, likewise resisting contraction–expansion in different solvents, for instance.
Collapse
|
3
|
Deus IA, Santos SC, Custódio CA, Mano JF. Designing highly customizable human based platforms for cell culture using proteins from the amniotic membrane. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112574. [PMID: 35525741 DOI: 10.1016/j.msec.2021.112574] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022]
Abstract
In the past few years researchers have witnessed a paradigm shift in the development of biomaterials for drug discovery, tissue engineering, and regenerative medicine. After the great advances resulting from the transition of the 2D to the 3D, the new focus has been to increase the clinical relevance of such systems, as well as avoid the use of animals, by developing platforms that better replicate the human physiology in vitro. In this sense, we envisage the use of human matrices extracted from ethically sourced and readily available tissues as an optimal and promising alternative to currently used approaches. Hereupon, we report for the first time the chemical modification of human ECM proteins from the amniotic membrane (AM) with photoresponsive groups to produce bioinks and hydrogel precursors to engineer customizable platforms that are representative of native tissues and capable of supporting long-term cell culture. Our results demonstrated an efficient decellularization, liquefaction and functionalization of AM-derived ECM with methacryloyl domains (AMMA), with production of stable and versatile hydrogels. Mechanical characterization evidenced an increased compression strength as a function of methacrylation degree and decellularized ECM concentration. Three-dimensional (3D) stem cell culture in the AMMA hydrogels resulted in viable and proliferative cells up to 7 days; moreover, the mouldable character of the hydrogel precursors permits the processing of patterned hydrogel constructs allowing the control over cellular alignment and elongation, or microgels with highly tunable shape.
Collapse
Affiliation(s)
- Inês A Deus
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Sara C Santos
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Catarina A Custódio
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
4
|
Bicho D, Canadas RF, Gonçalves C, Pina S, Reis RL, Oliveira JM. Porous aligned ZnSr-doped β-TCP/silk fibroin scaffolds using ice-templating method for bone tissue engineering applications. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1966-1982. [PMID: 34228590 DOI: 10.1080/09205063.2021.1952382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The bone is a complex and dynamic structure subjected to constant stress and remodeling. Due to the worldwide incidence of bone disorders, tissue scaffolds and engineered bone tissues have emerged as solutions for bone grafting, which require sophisticated scaffolding architectures while keeping high mechanical performance. However, the conjugation of a bone-like scaffold architecture with efficient mechanical properties is still a critical challenge for biomedical applications. In this sense, the present study focused on the modulating the architecture of silk fibroin (SF) scaffolds crosslinked with horseradish peroxidase and mixed with zinc (Zn) and strontium (Sr)-doped β-tricalcium phosphate (ZnSr.TCP) to mimic bone structures. The ZnSr.TCP-SF hydrogels were tuned by programmable ice-templating parameters, and further freeze-dried, in order to obtain 3D scaffolds with controlled pore orientation. The results showed interconnected channels in the ZnSr.TCP-SF scaffolds that mimic the porous network of the native subchondral bone matrix. The architecture of the scaffolds was characterized by microCT, showing tunable pore size according to freezing temperatures (-196 °C: ∼80.2 ± 20.5 µm; -80 °C: ∼73.1 ± 20.5 µm; -20 °C: ∼104.7 ± 33.7 µm). The swelling ratio, weight loss, and rheological properties were also assessed, revealing efficient scaffold integrity and morphology after aqueous immersion. Thus, the ZnSr.TCP-SF scaffolds made of aligned porous structure were developed as affordable candidates for future applications in clinical osteoregeneration and in vitro bone tissue modelling.
Collapse
Affiliation(s)
- D Bicho
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - R F Canadas
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - C Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - S Pina
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - R L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - J M Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
5
|
Soto F, Wang J, Deshmukh S, Demirci U. Reversible Design of Dynamic Assemblies at Small Scales. ADVANCED INTELLIGENT SYSTEMS (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 3:2000193. [PMID: 35663639 PMCID: PMC9165726 DOI: 10.1002/aisy.202000193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Indexed: 05/08/2023]
Abstract
Emerging bottom-up fabrication methods have enabled the assembly of synthetic colloids, microrobots, living cells, and organoids to create intricate structures with unique properties that transcend their individual components. This review provides an access point to the latest developments in externally driven assembly of synthetic and biological components. In particular, we emphasize reversibility, which enables the fabrication of multiscale systems that would not be possible under traditional techniques. Magnetic, acoustic, optical, and electric fields are the most promising methods for controlling the reversible assembly of biological and synthetic subunits since they can reprogram their assembly by switching on/off the external field or shaping these fields. We feature capabilities to dynamically actuate the assembly configuration by modulating the properties of the external stimuli, including frequency and amplitude. We describe the design principles which enable the assembly of reconfigurable structures. Finally, we foresee that the high degree of control capabilities offered by externally driven assembly will enable broad access to increasingly robust design principles towards building advanced dynamic intelligent systems.
Collapse
Affiliation(s)
- Fernando Soto
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, USA
| | - Jie Wang
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, USA
| | - Shreya Deshmukh
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, USA
- Department of Bioengineering, School of Engineering, School of Medicine, Stanford University, Stanford, California, 94305-4125, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, California 94304-5427, USA
| |
Collapse
|
6
|
Edelbrock AN, Clemons TD, Chin SM, Roan JJW, Bruckner EP, Álvarez Z, Edelbrock JF, Wek KS, Stupp SI. Superstructured Biomaterials Formed by Exchange Dynamics and Host-Guest Interactions in Supramolecular Polymers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004042. [PMID: 33898187 PMCID: PMC8061421 DOI: 10.1002/advs.202004042] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/21/2020] [Indexed: 05/12/2023]
Abstract
Dynamic and reversible assembly of molecules is ubiquitous in the hierarchical superstructures of living systems and plays a key role in cellular functions. Recent work from the laboratory reported on the reversible formation of such superstructures in systems of peptide amphiphiles conjugated to oligonucleotides and electrostatically complimentary peptide sequences. Here, a supramolecular system is reported upon where exchange dynamics and host-guest interactions between β-cyclodextrin and adamantane on peptide amphiphiles lead to superstructure formation. Superstructure formation with bundled nanoribbons generates a mechanically robust hydrogel with a highly porous architecture that can be 3D printed. Functionalization of the porous superstructured material with a biological signal results in a matrix with significant in vitro bioactivity toward neurons that could be used as a supramolecular model to design novel biomaterials.
Collapse
Affiliation(s)
- Alexandra N. Edelbrock
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIL60208USA
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
| | - Tristan D. Clemons
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
- Department of ChemistryNorthwestern UniversityEvanstonIL60208USA
| | - Stacey M. Chin
- Department of ChemistryNorthwestern UniversityEvanstonIL60208USA
| | - Joshua J. W. Roan
- Department of Materials Science and EngineeringNorthwestern UniversityEvanstonIL60208USA
| | - Eric P. Bruckner
- Department of Materials Science and EngineeringNorthwestern UniversityEvanstonIL60208USA
| | - Zaida Álvarez
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
- Department of MedicineNorthwestern UniversityChicagoIL60611USA
| | - Jack F. Edelbrock
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
- Department of Materials Science and EngineeringNorthwestern UniversityEvanstonIL60208USA
- Department of MedicineNorthwestern UniversityChicagoIL60611USA
| | - Kristen S. Wek
- Department of Materials Science and EngineeringNorthwestern UniversityEvanstonIL60208USA
| | - Samuel I. Stupp
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIL60208USA
- Simpson Querrey InstituteNorthwestern UniversityChicagoIL60611USA
- Department of ChemistryNorthwestern UniversityEvanstonIL60208USA
- Department of Materials Science and EngineeringNorthwestern UniversityEvanstonIL60208USA
- Department of MedicineNorthwestern UniversityChicagoIL60611USA
| |
Collapse
|
7
|
Zhu H, Yang H, Ma Y, Lu TJ, Xu F, Genin GM, Lin M. Spatiotemporally Controlled Photoresponsive Hydrogels: Design and Predictive Modeling from Processing through Application. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2000639. [PMID: 32802013 PMCID: PMC7418561 DOI: 10.1002/adfm.202000639] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/16/2020] [Indexed: 05/16/2023]
Abstract
Photoresponsive hydrogels (PRHs) are soft materials whose mechanical and chemical properties can be tuned spatially and temporally with relative ease. Both photo-crosslinkable and photodegradable hydrogels find utility in a range of biomedical applications that require tissue-like properties or programmable responses. Progress in engineering with PRHs is facilitated by the development of theoretical tools that enable optimization of their photochemistry, polymer matrices, nanofillers, and architecture. This review brings together models and design principles that enable key applications of PRHs in tissue engineering, drug delivery, and soft robotics, and highlights ongoing challenges in both modeling and application.
Collapse
Affiliation(s)
- Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Haiqian Yang
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical StructuresNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
- MOE Key Laboratory for Multifunctional Materials and StructuresXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Guy M. Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
- Department of Mechanical Engineering & Materials ScienceWashington University in St. LouisSt. LouisMO63130USA
- NSF Science and Technology Center for Engineering MechanobiologyWashington University in St. LouisSt. LouisMO63130USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| |
Collapse
|
8
|
Zhao Y, Demirci U, Chen Y, Chen P. Multiscale brain research on a microfluidic chip. LAB ON A CHIP 2020; 20:1531-1543. [PMID: 32150176 DOI: 10.1039/c9lc01010f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One major challenge in current brain research is generating an integrative understanding of the brain's functions and disorders from its multiscale neuronal architectures and connectivity. Thus, innovative neurotechnology tools are urgently required for deciphering the multiscale functional and structural organizations of the brain at hierarchical scales from the molecular to the organismal level by multiple brain research initiatives launched by the European Union, United States, Australia, Canada, China, Korea, and Japan. To meet this demand, microfluidic chips (μFCs) have rapidly evolved as a trans-scale neurotechnological toolset to enable multiscale studies of the brain due to their unique advantages in flexible microstructure design, multifunctional integration, accurate microenvironment control, and capacity for automatic sample processing. Here, we review the recent progress in applying innovative μFC-based neuro-technologies to promote multiscale brain research and uniquely focus on representative applications of μFCs to address challenges in brain research at each hierarchical level. We discuss the current trend of combinational applications of μFCs with other neuro- and biotechnologies, including optogenetics, brain organoids, and 3D bioprinting, for better multiscale brain research. In addition, we offer our insights into the existing outstanding questions at each hierarchical level of brain research that could potentially be addressed by advancing microfluidic techniques. This review will serve as a timely guide for bioengineers and neuroscientists to develop and apply μFC-based neuro-technologies for promoting basic and translational brain research.
Collapse
Affiliation(s)
- Yanan Zhao
- Department of Biomedical Engineering, Wuhan University School of Basic Medical Sciences, 115 Donghu Road, Wuhan 430071, China.
| | | | | | | |
Collapse
|
9
|
Dong J, Chen JF, Smalley M, Zhao M, Ke Z, Zhu Y, Tseng HR. Nanostructured Substrates for Detection and Characterization of Circulating Rare Cells: From Materials Research to Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903663. [PMID: 31566837 PMCID: PMC6946854 DOI: 10.1002/adma.201903663] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/02/2019] [Indexed: 05/03/2023]
Abstract
Circulating rare cells in the blood are of great significance for both materials research and clinical applications. For example, circulating tumor cells (CTCs) have been demonstrated as useful biomarkers for "liquid biopsy" of the tumor. Circulating fetal nucleated cells (CFNCs) have shown potential in noninvasive prenatal diagnostics. However, it is technically challenging to detect and isolate circulating rare cells due to their extremely low abundance compared to hematologic cells. Nanostructured substrates offer a unique solution to address these challenges by providing local topographic interactions to strengthen cell adhesion and large surface areas for grafting capture agents, resulting in improved cell capture efficiency, purity, sensitivity, and reproducibility. In addition, rare-cell retrieval strategies, including stimulus-responsiveness and additive reagent-triggered release on different nanostructured substrates, allow for on-demand retrieval of the captured CTCs/CFNCs with high cell viability and molecular integrity. Several nanostructured substrate-enabled CTC/CFNC assays are observed maturing from enumeration and subclassification to molecular analyses. These can one day become powerful tools in disease diagnosis, prognostic prediction, and dynamic monitoring of therapeutic response-paving the way for personalized medical care.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jie-Fu Chen
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
10
|
Ersoy F, Sokullu E. A compartmental 3D scaffold fabrication and alignment device for neurovascular co-culture and tri-culture. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab0dcc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
11
|
Seow WY, Kandasamy K, Purnamawati K, Sun W, Hauser CA. Thin peptide hydrogel membranes suitable as scaffolds for engineering layered biostructures. Acta Biomater 2019; 88:293-300. [PMID: 30721784 DOI: 10.1016/j.actbio.2019.02.001] [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: 07/16/2018] [Revised: 01/19/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
A short tetramer peptide, Ac-IVKC, spontaneously formed a hydrogel in water. Disulfide bonds were introduced via hydrogen peroxide (H2O2)-assisted oxidation, resulting in (Ac-IVKC)2 dimers. The extent of disulfide bond formation and gel stiffness increased with the amount of H2O2 used and 100% dimerization was achieved with 0.2% H2O2. The resultant gel achieved an elastic modulus of ∼0.9 MPa, which to our knowledge, has not been reported for peptide-based hydrogels. The enhanced mechanical property enabled the fabrication of thin and transparent membranes. The hydrogel could also be handled with forceps at mm thickness, greatly increasing its ease of physical manipulation. Excess H2O2 was removed and the membrane was then infused with cell culture media. Various cells, including primary human corneal stromal and epithelial cells, were seeded onto the hydrogel membrane and demonstrated to remain viable. Depending on the intended application, specific cell combination or membrane stacking order could be used to engineer layered biostructures. STATEMENT OF SIGNIFICANCE: A short tetramer peptide - Ac-IVKC - spontaneously formed a hydrogel in water and disulfide bonds were introduced via hydrogen peroxide (H2O2)-assisted oxidation. The extent of disulfide-bond formation and gel stiffness were modulated by the amount of H2O2. At maximum disulfide-bond formation, the hydrogel achieved an elastic modulus of ∼0.9 MPa, which to our knowledge, has not been reported for peptide-based hydrogels. The enhanced mechanical property enabled the fabrication of thin transparent membranes that can be physically manipulated at mm thickness. The gels also supported 3D cell growth, including primary human corneal stromal and epithelial cells. Depending on the intended application, specific combination of cells or individual membrane stacking order could be used to engineer layered biostructures.
Collapse
|
12
|
Ren T, Grosshäuser B, Sridhar K, Nieland TJF, Tocchio A, Schepers U, Demirci U. 3-D geometry and irregular connectivity dictate neuronal firing in frequency domain and synchronization. Biomaterials 2019; 197:171-181. [PMID: 30660993 DOI: 10.1016/j.biomaterials.2019.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 01/18/2023]
Abstract
The replication of the complex structure and three dimensional (3-D) interconnectivity of neurons in the brain is a great challenge. A few 3-D neuronal patterning approaches have been developed to mimic the cell distribution in the brain but none have demonstrated the relationship between 3-D neuron patterning and network connectivity. Here, we used photolithographic crosslinking to fabricate in vitro 3-D neuronal structures with distinct sizes, shapes or interconnectivities, i.e., milli-blocks, micro-stripes, separated micro-blocks and connected micro-blocks, which have spatial confinement from "Z" dimension to "XYZ" dimension. During a 4-week culture period, the 3-D neuronal system has shown high cell viability, axonal, dendritic, synaptic growth and neural network activity of cortical neurons. We further studied the calcium oscillation of neurons in different 3-D patterns and used signal processing both in Fast Fourier Transform (FFT) and time domain (TD) to model the fluorescent signal variation. We observed that the firing frequency decreased as the spatial confinement in 3-D system increased. Besides, the neuronal synchronization significantly decreased by irregularly connecting micro-blocks, indicating that network connectivity can be adjusted by changing the linking conditions of 3-D gels. Earlier works showed the importance of 3-D culture over 2-D in terms of cell growth. Here, we showed that not only 3-D geometry over 2-D culture matters, but also the spatial organization of cells in 3-D dictates the neuronal firing frequency and synchronicity.
Collapse
Affiliation(s)
- Tanchen Ren
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Bianka Grosshäuser
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA; Institute of Toxicology and Gentics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz, Eggenstein-Leopoldshafen, 76344, Germany
| | - Kaushik Sridhar
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Thomas J F Nieland
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Alessandro Tocchio
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA
| | - Ute Schepers
- Institute of Toxicology and Gentics, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz, Eggenstein-Leopoldshafen, 76344, Germany
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA, 94304, USA.
| |
Collapse
|
13
|
Obst F, Simon D, Mehner PJ, Neubauer JW, Beck A, Stroyuk O, Richter A, Voit B, Appelhans D. One-step photostructuring of multiple hydrogel arrays for compartmentalized enzyme reactions in microfluidic devices. REACT CHEM ENG 2019. [DOI: 10.1039/c9re00349e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A moulding technique is presented for the simultaneous photostructuring on the μm scale of hydrogels with nanomaterials on one substrate, usable for the fabrication of microfluidic double-chamber reactors.
Collapse
Affiliation(s)
- Franziska Obst
- Leibniz-Institut für Polymerforschung Dresden e.V
- 01069 Dresden
- Germany
- Technische Universität Dresden
- Organische Chemie der Polymere
| | - David Simon
- Leibniz-Institut für Polymerforschung Dresden e.V
- 01069 Dresden
- Germany
- Technische Universität Dresden
- Organische Chemie der Polymere
| | - Philipp J. Mehner
- Technische Universität Dresden
- Institut für Halbleiter- und Mikrosystemtechnik
- 01187 Dresden
- Germany
| | - Jens W. Neubauer
- Leibniz-Institut für Polymerforschung Dresden e.V
- 01069 Dresden
- Germany
| | - Anthony Beck
- Technische Universität Dresden
- Institut für Halbleiter- und Mikrosystemtechnik
- 01187 Dresden
- Germany
| | - Oleksandr Stroyuk
- Technische Universität Dresden
- Physikalische Chemie
- 01069 Dresden
- Germany
| | - Andreas Richter
- Technische Universität Dresden
- Institut für Halbleiter- und Mikrosystemtechnik
- 01187 Dresden
- Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V
- 01069 Dresden
- Germany
- Technische Universität Dresden
- Organische Chemie der Polymere
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V
- 01069 Dresden
- Germany
| |
Collapse
|
14
|
Canadas RF, Ren T, Tocchio A, Marques AP, Oliveira JM, Reis RL, Demirci U. Tunable anisotropic networks for 3-D oriented neural tissue models. Biomaterials 2018; 181:402-414. [DOI: 10.1016/j.biomaterials.2018.07.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 07/24/2018] [Accepted: 07/28/2018] [Indexed: 02/06/2023]
|
15
|
Magariños AM, Pedron S, Creixell M, Kilinc M, Tabansky I, Pfaff DW, Harley BAC. The Feasibility of Encapsulated Embryonic Medullary Reticular Cells to Grow and Differentiate Into Neurons in Functionalized Gelatin-Based Hydrogels. FRONTIERS IN MATERIALS 2018; 5:40. [PMID: 30687706 PMCID: PMC6345411 DOI: 10.3389/fmats.2018.00040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The study of the behavior of embryonic neurons in controlled in vitro conditions require methodologies that take advantage of advanced tissue engineering approaches to replicate elements of the developing brain extracellular matrix. We report here a series of experiments that explore the potential of photo-polymerized gelatin hydrogels to culture primary embryonic neurons. We employed large medullary reticular neurons whose activity is essential for brain arousal as well as a library of gelatin hydrogels that span a range of mechanical properties, inclusion of brain-mimetic hyaluronic acid, and adhesion peptides. These hydrogel platforms showed inherent capabilities to sustain neuronal viability and were permissive for neuronal differentiation, resulting in the development of neurite outgrowth under specific conditions. The maturation of embryonic medullary reticular cells took place in the absence of growth factors or other exogenous bioactive molecules. Immunocytochemistry labeling of neuron-specific tubulin confirmed the initiation of neural differentiation. Thus, this methodology provides an important validation for future studies of nerve cell growth and maintenance.
Collapse
Affiliation(s)
- Ana M. Magariños
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Sara Pedron
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Marc Creixell
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Murat Kilinc
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Inna Tabansky
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Donald W. Pfaff
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY, United States
| | - Brendan A. C. Harley
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
16
|
Rose JC, De Laporte L. Hierarchical Design of Tissue Regenerative Constructs. Adv Healthc Mater 2018; 7:e1701067. [PMID: 29369541 DOI: 10.1002/adhm.201701067] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/01/2017] [Indexed: 02/05/2023]
Abstract
The worldwide shortage of organs fosters significant advancements in regenerative therapies. Tissue engineering and regeneration aim to supply or repair organs or tissues by combining material scaffolds, biochemical signals, and cells. The greatest challenge entails the creation of a suitable implantable or injectable 3D macroenvironment and microenvironment to allow for ex vivo or in vivo cell-induced tissue formation. This review gives an overview of the essential components of tissue regenerating scaffolds, ranging from the molecular to the macroscopic scale in a hierarchical manner. Further, this review elaborates about recent pivotal technologies, such as photopatterning, electrospinning, 3D bioprinting, or the assembly of micrometer-scale building blocks, which enable the incorporation of local heterogeneities, similar to most native extracellular matrices. These methods are applied to mimic a vast number of different tissues, including cartilage, bone, nerves, muscle, heart, and blood vessels. Despite the tremendous progress that has been made in the last decade, it remains a hurdle to build biomaterial constructs in vitro or in vivo with a native-like structure and architecture, including spatiotemporal control of biofunctional domains and mechanical properties. New chemistries and assembly methods in water will be crucial to develop therapies that are clinically translatable and can evolve into organized and functional tissues.
Collapse
Affiliation(s)
- Jonas C. Rose
- DWI—Leibniz Institute for Interactive Materials Forckenbeckstr. 50 Aachen D‐52074 Germany
| | - Laura De Laporte
- DWI—Leibniz Institute for Interactive Materials Forckenbeckstr. 50 Aachen D‐52074 Germany
| |
Collapse
|
17
|
Song H, Cui J, Simonyi A, Johnson CE, Hubler GK, DePalma RG, Gu Z. Linking blast physics to biological outcomes in mild traumatic brain injury: Narrative review and preliminary report of an open-field blast model. Behav Brain Res 2018; 340:147-158. [DOI: 10.1016/j.bbr.2016.08.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 08/13/2016] [Accepted: 08/19/2016] [Indexed: 12/14/2022]
|
18
|
Hogan LE, Vasquez J, Hobbs KS, Hanhauser E, Aguilar-Rodriguez B, Hussien R, Thanh C, Gibson EA, Carvidi AB, Smith LCB, Khan S, Trapecar M, Sanjabi S, Somsouk M, Stoddart CA, Kuritzkes DR, Deeks SG, Henrich TJ. Increased HIV-1 transcriptional activity and infectious burden in peripheral blood and gut-associated CD4+ T cells expressing CD30. PLoS Pathog 2018; 14:e1006856. [PMID: 29470552 PMCID: PMC5823470 DOI: 10.1371/journal.ppat.1006856] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022] Open
Abstract
HIV-1-infected cells persist indefinitely despite the use of combination antiretroviral therapy (ART), and novel therapeutic strategies to target and purge residual infected cells in individuals on ART are urgently needed. Here, we demonstrate that CD4+ T cell-associated HIV-1 RNA is often highly enriched in cells expressing CD30, and that cells expressing this marker considerably contribute to the total pool of transcriptionally active CD4+ lymphocytes in individuals on suppressive ART. Using in situ RNA hybridization studies, we show co-localization of CD30 with HIV-1 transcriptional activity in gut-associated lymphoid tissues. We also demonstrate that ex vivo treatment with brentuximab vedotin, an antibody-drug conjugate (ADC) that targets CD30, significantly reduces the total amount of HIV-1 DNA in peripheral blood mononuclear cells obtained from infected, ART-suppressed individuals. Finally, we observed that an HIV-1-infected individual, who received repeated brentuximab vedotin infusions for lymphoma, had no detectable virus in peripheral blood mononuclear cells. Overall, CD30 may be a marker of residual, transcriptionally active HIV-1 infected cells in the setting of suppressive ART. Given that CD30 is only expressed on a small number of total mononuclear cells, it is a potential therapeutic target of persistent HIV-1 infection. Previous studies have shown that higher levels of soluble CD30 are associated with HIV-1 disease progression. Many of these studies, however, were performed prior to the implementation of combination ART, and the relationship between surface CD30 expression, soluble CD30 and HIV-1 infection in ART suppressed individuals, or those with viremic control off ART, is not known. We demonstrate that cell-associated HIV-1 RNA is highly enriched in CD4+ T cells expressing CD30, a member of the tumor necrosis factor receptor superfamily. These findings were observed in several HIV-1 infected donor groups, regardless of whether or not the participants were receiving suppressive ART. Furthermore, we demonstrate that ex vivo treatment with brentuximab vedotin, an antibody-drug conjugate that targets CD30, reduces the total amount of HIV-1 DNA in PBMC obtained from infected individuals. Finally, we show through in situ RNA hybridization studies that CD30 and HIV transcriptional activity co-localize in cells from gut biopsies obtained from HIV-1 infected donors. These data suggest that CD30 may be a marker of residual, transcriptionally active HIV-1 infected cells in the setting of suppressive ART.
Collapse
Affiliation(s)
- Louise E. Hogan
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (LEH); (TJH)
| | - Joshua Vasquez
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Kristen S. Hobbs
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Emily Hanhauser
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Brandon Aguilar-Rodriguez
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Rajaa Hussien
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Cassandra Thanh
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Erica A. Gibson
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Alexander B. Carvidi
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Louis C. B. Smith
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Shahzada Khan
- Virology and Immunology, Gladstone Institutes, San Francisco, California, United States of America
| | - Martin Trapecar
- Virology and Immunology, Gladstone Institutes, San Francisco, California, United States of America
| | - Shomyseh Sanjabi
- Virology and Immunology, Gladstone Institutes, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Ma Somsouk
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Cheryl A. Stoddart
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Daniel R. Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Steven G. Deeks
- Positive Health Program, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (LEH); (TJH)
| |
Collapse
|
19
|
|
20
|
Zhang W, Huang G, Ng K, Ji Y, Gao B, Huang L, Zhou J, Lu TJ, Xu F. Engineering ellipsoidal cap-like hydrogel particles as building blocks or sacrificial templates for three-dimensional cell culture. Biomater Sci 2018; 6:885-892. [DOI: 10.1039/c7bm01186e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AAO template-assisted fabrication of ellipsoidal cap-like hydrogel particles as building blocks or sacrificial templates for 3D cell culture.
Collapse
Affiliation(s)
- Weiwei Zhang
- Non-equilibrium Condensed Matter and Quantum Engineering Laboratory
- The Key Laboratory of Ministry of Education
- School of Science
- Xi'an Jiaotong University
- Xi'an 710049
| | - Guoyou Huang
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| | - Kelvin Ng
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
- Department of Biomedical Engineering
| | - Yuan Ji
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| | - Bin Gao
- Department of Endocrinology and Metabolism
- Xijing Hospital
- Fourth Military Medical University
- Xi'an 710054
- P.R. China
| | - Liqing Huang
- Non-equilibrium Condensed Matter and Quantum Engineering Laboratory
- The Key Laboratory of Ministry of Education
- School of Science
- Xi'an Jiaotong University
- Xi'an 710049
| | - Jinxiong Zhou
- State Key Laboratory for Strength and Vibration of Mechanical Structures
- School of Aerospace
- Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
- MOE Key Laboratory for Multifunctional Materials and Structures
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| |
Collapse
|
21
|
Huang G, Li F, Zhao X, Ma Y, Li Y, Lin M, Jin G, Lu TJ, Genin GM, Xu F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem Rev 2017; 117:12764-12850. [PMID: 28991456 PMCID: PMC6494624 DOI: 10.1021/acs.chemrev.7b00094] [Citation(s) in RCA: 514] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell microenvironment has emerged as a key determinant of cell behavior and function in development, physiology, and pathophysiology. The extracellular matrix (ECM) within the cell microenvironment serves not only as a structural foundation for cells but also as a source of three-dimensional (3D) biochemical and biophysical cues that trigger and regulate cell behaviors. Increasing evidence suggests that the 3D character of the microenvironment is required for development of many critical cell responses observed in vivo, fueling a surge in the development of functional and biomimetic materials for engineering the 3D cell microenvironment. Progress in the design of such materials has improved control of cell behaviors in 3D and advanced the fields of tissue regeneration, in vitro tissue models, large-scale cell differentiation, immunotherapy, and gene therapy. However, the field is still in its infancy, and discoveries about the nature of cell-microenvironment interactions continue to overturn much early progress in the field. Key challenges continue to be dissecting the roles of chemistry, structure, mechanics, and electrophysiology in the cell microenvironment, and understanding and harnessing the roles of periodicity and drift in these factors. This review encapsulates where recent advances appear to leave the ever-shifting state of the art, and it highlights areas in which substantial potential and uncertainty remain.
Collapse
Affiliation(s)
- Guoyou Huang
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Chemistry, School of Science,
Xi’an Jiaotong University, Xi’an 710049, People’s Republic
of China
| | - Xin Zhao
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Interdisciplinary Division of Biomedical
Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong,
People’s Republic of China
| | - Yufei Ma
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Yuhui Li
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Min Lin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Guorui Jin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- MOE Key Laboratory for Multifunctional Materials
and Structures, Xi’an Jiaotong University, Xi’an 710049,
People’s Republic of China
| | - Guy M. Genin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Mechanical Engineering &
Materials Science, Washington University in St. Louis, St. Louis 63130, MO,
USA
- NSF Science and Technology Center for
Engineering MechanoBiology, Washington University in St. Louis, St. Louis 63130,
MO, USA
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| |
Collapse
|
22
|
Guan X, Avci-Adali M, Alarçin E, Cheng H, Kashaf SS, Li Y, Chawla A, Jang HL, Khademhosseini A. Development of hydrogels for regenerative engineering. Biotechnol J 2017; 12:10.1002/biot.201600394. [PMID: 28220995 PMCID: PMC5503693 DOI: 10.1002/biot.201600394] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 11/07/2022]
Abstract
The aim of regenerative engineering is to restore complex tissues and biological systems through convergence in the fields of advanced biomaterials, stem cell science, and developmental biology. Hydrogels are one of the most attractive biomaterials for regenerative engineering, since they can be engineered into tissue mimetic 3D scaffolds to support cell growth due to their similarity to native extracellular matrix. Advanced nano- and micro-technologies have dramatically increased the ability to control properties and functionalities of hydrogel materials by facilitating biomimetic fabrication of more sophisticated compositions and architectures, thus extending our understanding of cell-matrix interactions at the nanoscale. With this perspective, this review discusses the most commonly used hydrogel materials and their fabrication strategies for regenerative engineering. We highlight the physical, chemical, and functional modulation of hydrogels to design and engineer biomimetic tissues based on recent achievements in nano- and micro-technologies. In addition, current hydrogel-based regenerative engineering strategies for treating multiple tissues, such as musculoskeletal, nervous and cardiac tissue, are also covered in this review. The interaction of multiple disciplines including materials science, cell biology, and chemistry, will further play an important role in the design of functional hydrogels for the regeneration of complex tissues.
Collapse
Affiliation(s)
- Xiaofei Guan
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Orthopedic Department, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstr. 7/1, Tuebingen 72076, Germany
| | - Emine Alarçin
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul 34668, Turkey
| | - Hao Cheng
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sara Saheb Kashaf
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuxiao Li
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aditya Chawla
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hae Lin Jang
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience & Technology, Konkuk University, Seoul 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
23
|
Serpooshan V, Chen P, Wu H, Lee S, Sharma A, Hu DA, Venkatraman S, Ganesan AV, Usta OB, Yarmush M, Yang F, Wu JC, Demirci U, Wu SM. Bioacoustic-enabled patterning of human iPSC-derived cardiomyocytes into 3D cardiac tissue. Biomaterials 2017; 131:47-57. [PMID: 28376365 DOI: 10.1016/j.biomaterials.2017.03.037] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/05/2017] [Accepted: 03/23/2017] [Indexed: 12/16/2022]
Abstract
The creation of physiologically-relevant human cardiac tissue with defined cell structure and function is essential for a wide variety of therapeutic, diagnostic, and drug screening applications. Here we report a new scalable method using Faraday waves to enable rapid aggregation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) into predefined 3D constructs. At packing densities that approximate native myocardium (108-109 cells/ml), these hiPSC-CM-derived 3D tissues demonstrate significantly improved cell viability, metabolic activity, and intercellular connection when compared to constructs with random cell distribution. Moreover, the patterned hiPSC-CMs within the constructs exhibit significantly greater levels of contractile stress, beat frequency, and contraction-relaxation rates, suggesting their improved maturation. Our results demonstrate a novel application of Faraday waves to create stem cell-derived 3D cardiac tissue that resembles the cellular architecture of a native heart tissue for diverse basic research and clinical applications.
Collapse
Affiliation(s)
- Vahid Serpooshan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Pu Chen
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, Stanford University School of Medicine, Canary Center for Early Cancer Detection, Stanford, CA, USA; Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China; Institute of Model Animal of Wuhan University, Wuhan, China
| | - Haodi Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Soah Lee
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Arun Sharma
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel A Hu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sneha Venkatraman
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Biology Program, California State University Channel Islands, Camarillo, CA, USA
| | | | - Osman Berk Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston 02144, MA, USA
| | - Martin Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston 02144, MA, USA; Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway 08854, NJ, USA
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Department of Radiology, Stanford University School of Medicine, Canary Center for Early Cancer Detection, Stanford, CA, USA; Department of Electrical Engineering (by Courtesy), Stanford University School of Engineering, Stanford, CA, USA.
| | - Sean M Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
24
|
Albritton JL, Miller JS. 3D bioprinting: improving in vitro models of metastasis with heterogeneous tumor microenvironments. Dis Model Mech 2017; 10:3-14. [PMID: 28067628 PMCID: PMC5278522 DOI: 10.1242/dmm.025049] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Even with many advances in treatment over the past decades, cancer still remains a leading cause of death worldwide. Despite the recognized relationship between metastasis and increased mortality rate, surprisingly little is known about the exact mechanism of metastatic progression. Currently available in vitro models cannot replicate the three-dimensionality and heterogeneity of the tumor microenvironment sufficiently to recapitulate many of the known characteristics of tumors in vivo Our understanding of metastatic progression would thus be boosted by the development of in vitro models that could more completely capture the salient features of cancer biology. Bioengineering groups have been working for over two decades to create in vitro microenvironments for application in regenerative medicine and tissue engineering. Over this time, advances in 3D printing technology and biomaterials research have jointly led to the creation of 3D bioprinting, which has improved our ability to develop in vitro models with complexity approaching that of the in vivo tumor microenvironment. In this Review, we give an overview of 3D bioprinting methods developed for tissue engineering, which can be directly applied to constructing in vitro models of heterogeneous tumor microenvironments. We discuss considerations and limitations associated with 3D printing and highlight how these advances could be harnessed to better model metastasis and potentially guide the development of anti-cancer strategies.
Collapse
Affiliation(s)
- Jacob L Albritton
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Jordan S Miller
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| |
Collapse
|
25
|
3-D Microwell Array System for Culturing Virus Infected Tumor Cells. Sci Rep 2016; 6:39144. [PMID: 28004818 PMCID: PMC5177905 DOI: 10.1038/srep39144] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 11/18/2016] [Indexed: 12/18/2022] Open
Abstract
Cancer cells have been increasingly grown in pharmaceutical research to understand tumorigenesis and develop new therapeutic drugs. Currently, cells are typically grown using two-dimensional (2-D) cell culture approaches, where the native tumor microenvironment is difficult to recapitulate. Thus, one of the main obstacles in oncology is the lack of proper infection models that recount main features present in tumors. In recent years, microtechnology-based platforms have been employed to generate three-dimensional (3-D) models that better mimic the native microenvironment in cell culture. Here, we present an innovative approach to culture Kaposi’s sarcoma-associated herpesvirus (KSHV) infected human B cells in 3-D using a microwell array system. The results demonstrate that the KSHV-infected B cells can be grown up to 15 days in a 3-D culture. Compared with 2-D, cells grown in 3-D had increased numbers of KSHV latency-associated nuclear antigen (LANA) dots, as detected by immunofluorescence microscopy, indicating a higher viral genome copy number. Cells in 3-D also demonstrated a higher rate of lytic reactivation. The 3-D microwell array system has the potential to improve 3-D cell oncology models and allow for better-controlled studies for drug discovery.
Collapse
|
26
|
Hasturk O, Sivas A, Karasozen B, Demirci U, Hasirci N, Hasirci V. Quantification of Type, Timing, and Extent of Cell Body and Nucleus Deformations Caused by the Dimensions and Hydrophilicity of Square Prism Micropillars. Adv Healthc Mater 2016; 5:2972-2982. [PMID: 27925459 DOI: 10.1002/adhm.201600857] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/21/2016] [Indexed: 01/30/2023]
Abstract
Novel digital analysis strategies are developed for the quantification of changes in the cytoskeletal and nuclear morphologies of mesenchymal stem cells cultured on micropillars. Severe deformations of nucleus and distinct conformational changes of cell body ranging from extensive elongation to branching are visualized and quantified. These deformations are caused mainly by the dimensions and hydrophilicity of the micropillars.
Collapse
Affiliation(s)
- Onur Hasturk
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Abdullah Sivas
- Institute of Applied Mathematics; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Bulent Karasozen
- Institute of Applied Mathematics; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Utkan Demirci
- Bio-Acoustic-MEMs in Medicine (BAMM) Laboratory; Stanford School of Medicine; Palo Alto CA 94394 USA
| | - Nesrin Hasirci
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
- Department of Chemistry; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Vasif Hasirci
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
- Department of Biological Sciences; Middle East Technical University (METU); Ankara 06800 Turkey
| |
Collapse
|
27
|
Hou K, Wang H, Lin Y, Chen S, Yang S, Cheng Y, Hsiao BS, Zhu M. Large Scale Production of Continuous Hydrogel Fibers with Anisotropic Swelling Behavior by Dynamic-Crosslinking-Spinning. Macromol Rapid Commun 2016; 37:1795-1801. [DOI: 10.1002/marc.201600430] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/29/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Kai Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| | - Huiyi Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| | - Yunyin Lin
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| | - Shaohua Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| | - Shengyuan Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| | - Yanhua Cheng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| | - Benjamin S. Hsiao
- Department of Chemistry; Stony Brook University; Stony Brook NY 11794 USA
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; College of Materials Science and Engineering; Donghua University; 2999 North Renmin Road Shanghai 201620 P. R. China
| |
Collapse
|
28
|
Morgan KY, Sklaviadis D, Tochka ZL, Fischer KM, Hearon K, Morgan TD, Langer R, Freed LE. Multi-Material Tissue Engineering Scaffold with Hierarchical Pore Architecture. ADVANCED FUNCTIONAL MATERIALS 2016; 26:5873-5883. [PMID: 27942257 PMCID: PMC5142531 DOI: 10.1002/adfm.201601146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Multi-material polymer scaffolds with multiscale pore architectures were characterized and tested with vascular and heart cells as part of a platform for replacing damaged heart muscle. Vascular and muscle scaffolds were constructed from a new material, poly(limonene thioether) (PLT32i), which met the design criteria of slow biodegradability, elastomeric mechanical properties, and facile processing. The vascular-parenchymal interface was a poly(glycerol sebacate) (PGS) porous membrane that met different criteria of rapid biodegradability, high oxygen permeance, and high porosity. A hierarchical architecture of primary (macroscale) and secondary (microscale) pores was created by casting the PLT32i prepolymer onto sintered spheres of poly(methyl methacrylate) (PMMA) within precisely patterned molds followed by photocuring, de-molding, and leaching out the PMMA. Pre-fabricated polymer templates were cellularized, assembled, and perfused in order to engineer spatially organized, contractile heart tissue. Structural and functional analyses showed that the primary pores guided heart cell alignment and enabled robust perfusion while the secondary pores increased heart cell retention and reduced polymer volume fraction.
Collapse
Affiliation(s)
- Kathy Ye Morgan
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Demetra Sklaviadis
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zachary L. Tochka
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kristin M. Fischer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Keith Hearon
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thomas D. Morgan
- Harvard University School of Engineering & Applied Science, Cambridge, MA 02138, USA
| | - Robert Langer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lisa E. Freed
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Materials Engineering Division, Draper, Cambridge, MA 02139, USA
| |
Collapse
|
29
|
Ma C, Tian C, Zhao L, Wang J. Pneumatic-aided micro-molding for flexible fabrication of homogeneous and heterogeneous cell-laden microgels. LAB ON A CHIP 2016; 16:2609-2617. [PMID: 27229899 DOI: 10.1039/c6lc00540c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Microgels are favorable for numerous applications such as drug delivery, biomaterials science and tissue engineering. Conventionally, photolithographic methods and micro-molding techniques are extensively exploited to prepare microgels; however, they are, respectively, limited to photocrosslinkable polymers and inadequate to generate serially patterned hydrogels due to the static nature of utilized molds. Herein, we proposed a simple and versatile approach, termed pneumatic-aided micro-molding (PAM), to flexibly fabricate microgels with precise control over multiple cell types and microarchitectures of hydrogels through strategically designed pneumatic microvalves. Using the PAM approach, different cells were encapsulated in various hydrogels that had well-defined geometries. Additionally, single/multiple micro-channeled cell-laden microgels were fabricated, of which the shape, number and arrangement could be finely tuned by varying microvalve configurations. Moreover, multi-compartmental microgels comprising composite hydrogel structures were engineered following a two-step PAM, which demonstrated the utility for biomimetically constructing a three-dimensional (3D) liver microtissue composed of a radially orchestrated network of hepatic cords and sinusoids. The resulting microtissue resembled the organizational complexity of the liver lobule and was applied for the evaluation of acetaminophen-induced hepatotoxicity. Collectively, the PAM strategy could be a useful and powerful tool in biomedical engineering, in vitro 3D cell culture, and fundamental biological studies.
Collapse
Affiliation(s)
- Chao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Chang Tian
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Lei Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Jinyi Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China. and College of Science, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
30
|
Abstract
The ability to three-dimensionally interweave biological and functional materials could enable the creation of bionic devices possessing unique and compelling geometries, properties, and functionalities. Indeed, interfacing high performance active devices with biology could impact a variety of fields, including regenerative bioelectronic medicines, smart prosthetics, medical robotics, and human-machine interfaces. Biology, from the molecular scale of DNA and proteins, to the macroscopic scale of tissues and organs, is three-dimensional, often soft and stretchable, and temperature sensitive. This renders most biological platforms incompatible with the fabrication and materials processing methods that have been developed and optimized for functional electronics, which are typically planar, rigid and brittle. A number of strategies have been developed to overcome these dichotomies. One particularly novel approach is the use of extrusion-based multi-material 3D printing, which is an additive manufacturing technology that offers a freeform fabrication strategy. This approach addresses the dichotomies presented above by (1) using 3D printing and imaging for customized, hierarchical, and interwoven device architectures; (2) employing nanotechnology as an enabling route for introducing high performance materials, with the potential for exhibiting properties not found in the bulk; and (3) 3D printing a range of soft and nanoscale materials to enable the integration of a diverse palette of high quality functional nanomaterials with biology. Further, 3D printing is a multi-scale platform, allowing for the incorporation of functional nanoscale inks, the printing of microscale features, and ultimately the creation of macroscale devices. This blending of 3D printing, novel nanomaterial properties, and 'living' platforms may enable next-generation bionic systems. In this review, we highlight this synergistic integration of the unique properties of nanomaterials with the versatility of extrusion-based 3D printing technologies to interweave nanomaterials and fabricate novel bionic devices.
Collapse
Affiliation(s)
- Yong Lin Kong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Maneesh K. Gupta
- Air Force Research Laboratories, Wright-Patterson Air Force Base, Ohio 45433, USA
| | - Blake N. Johnson
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Michael C. McAlpine
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
31
|
Yanagawa F, Sugiura S, Kanamori T. Hydrogel microfabrication technology toward three dimensional tissue engineering. Regen Ther 2016; 3:45-57. [PMID: 31245472 PMCID: PMC6581842 DOI: 10.1016/j.reth.2016.02.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 02/07/2023] Open
Abstract
The development of biologically relevant three-dimensional (3D) tissue constructs is essential for the alternative methods of organ transplantation in regenerative medicine, as well as the development of improved drug discovery assays. Recent technological advances in hydrogel microfabrication, such as micromolding, 3D bioprinting, photolithography, and stereolithography, have led to the production of 3D tissue constructs that exhibit biological functions with precise 3D microstructures. Furthermore, microfluidics technology has enabled the development of the perfusion culture of 3D tissue constructs with vascular networks. In this review, we present these hydrogel microfabrication technologies for the in vitro reconstruction and cultivation of 3D tissues. Additionally, we discuss current challenges and future perspectives of 3D tissue engineering.
Collapse
Affiliation(s)
- Fumiki Yanagawa
- Drug Assay Device Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5th, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Shinji Sugiura
- Drug Assay Device Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5th, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Toshiyuki Kanamori
- Drug Assay Device Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5th, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| |
Collapse
|
32
|
Namkoong B, Güven S, Ramesan S, Liaudanskaya V, Abzhanov A, Demirci U. Recapitulating cranial osteogenesis with neural crest cells in 3-D microenvironments. Acta Biomater 2016; 31:301-311. [PMID: 26675129 DOI: 10.1016/j.actbio.2015.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 11/11/2015] [Accepted: 12/02/2015] [Indexed: 01/20/2023]
Abstract
The experimental systems that recapitulate the complexity of native tissues and enable precise control over the microenvironment are becoming essential for the pre-clinical tests of therapeutics and tissue engineering. Here, we described a strategy to develop an in vitro platform to study the developmental biology of craniofacial osteogenesis. In this study, we directly osteo-differentiated cranial neural crest cells (CNCCs) in a 3-D in vitro bioengineered microenvironment. Cells were encapsulated in the gelatin-based photo-crosslinkable hydrogel and cultured up to three weeks. We demonstrated that this platform allows efficient differentiation of p75 positive CNCCs to cells expressing osteogenic markers corresponding to the sequential developmental phases of intramembranous ossification. During the course of culture, we observed a decrease in the expression of early osteogenic marker Runx2, while the other mature osteoblast and osteocyte markers such as Osterix, Osteocalcin, Osteopontin and Bone sialoprotein increased. We analyzed the ossification of the secreted matrix with alkaline phosphatase and quantified the newly secreted hydroxyapatite. The Field Emission Scanning Electron Microscope (FESEM) images of the bioengineered hydrogel constructs revealed the native-like osteocytes, mature osteoblasts, and cranial bone tissue morphologies with canaliculus-like intercellular connections. This platform provides a broadly applicable model system to potentially study diseases involving primarily embryonic craniofacial bone disorders, where direct diagnosis and adequate animal disease models are limited.
Collapse
Affiliation(s)
- Bumjin Namkoong
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sinan Güven
- Demirci BAMM Labs, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, Department of Electrical Engineering (By courtesy), Stanford School of Medicine, Palo Alto, CA 94304, USA; Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balcova, 35350 Izmir, Turkey
| | - Shwathy Ramesan
- Demirci BAMM Labs, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Volha Liaudanskaya
- Demirci BAMM Labs, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, Department of Electrical Engineering (By courtesy), Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Arhat Abzhanov
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Current address: Department of Life Sciences, Imperial College London, Silwood Park Campus Buckhurst Road, Ascot, Berkshire SL5 7PY, United Kingdom; Current address: Natural History Museum, Cromwell Road, London SW7 5BD, United Kingdom.
| | - Utkan Demirci
- Demirci BAMM Labs, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, Department of Electrical Engineering (By courtesy), Stanford School of Medicine, Palo Alto, CA 94304, USA; Demirci BAMM Labs, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Gaharwar AK, Arpanaei A, Andresen TL, Dolatshahi-Pirouz A. 3D Biomaterial Microarrays for Regenerative Medicine: Current State-of-the-Art, Emerging Directions and Future Trends. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:771-781. [PMID: 26607415 DOI: 10.1002/adma.201503918] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 08/19/2015] [Indexed: 06/05/2023]
Abstract
Three dimensional (3D) biomaterial microarrays hold enormous promise for regenerative medicine because of their ability to accelerate the design and fabrication of biomimetic materials. Such tissue-like biomaterials can provide an appropriate microenvironment for stimulating and controlling stem cell differentiation into tissue-specific lineages. The use of 3D biomaterial microarrays can, if optimized correctly, result in a more than 1000-fold reduction in biomaterials and cells consumption when engineering optimal materials combinations, which makes these miniaturized systems very attractive for tissue engineering and drug screening applications.
Collapse
Affiliation(s)
- Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Ayyoob Arpanaei
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Thomas L Andresen
- Technical University of Denmark, DTU Nanotech, Center for Nanomedicine and Theranostics, 2800, Kgs, Denmark
| | - Alireza Dolatshahi-Pirouz
- Technical University of Denmark, DTU Nanotech, Center for Nanomedicine and Theranostics, 2800, Kgs, Denmark
| |
Collapse
|
34
|
Bouyer C, Chen P, Güven S, Demirtaş TT, Nieland TJF, Padilla F, Demirci U. A Bio-Acoustic Levitational (BAL) Assembly Method for Engineering of Multilayered, 3D Brain-Like Constructs, Using Human Embryonic Stem Cell Derived Neuro-Progenitors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:161-7. [PMID: 26554659 DOI: 10.1002/adma.201503916] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/14/2015] [Indexed: 05/27/2023]
Abstract
A bio-acoustic levitational assembly method for engineering of multilayered, 3D brainlike constructs is presented. Acoustic radiation forces are used to levitate neuroprogenitors derived from human embryonic stem cells in 3D multilayered fibrin tissue constructs. The neuro-progenitor cells are subsequently differentiated in neural cells, resulting in a 3D neuronal construct with inter and intralayer neurite elongations.
Collapse
Affiliation(s)
- Charlène Bouyer
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Inserm, U1032, LabTau, University of Lyon, Lyon, F-69003, France
- LabEx DEVweCAN, University of Lyon, Lyon, F-69003, France
| | - Pu Chen
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Sinan Güven
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Tuğrul Tolga Demirtaş
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Thomas J F Nieland
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Frédéric Padilla
- Inserm, U1032, LabTau, University of Lyon, Lyon, F-69003, France
- LabEx DEVweCAN, University of Lyon, Lyon, F-69003, France
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Electrical Engineering (By Courtesy), Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
35
|
Chwalek K, Sood D, Cantley WL, White JD, Tang-Schomer M, Kaplan DL. Engineered 3D Silk-collagen-based Model of Polarized Neural Tissue. J Vis Exp 2015:e52970. [PMID: 26555926 PMCID: PMC4692668 DOI: 10.3791/52970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Despite huge efforts to decipher the anatomy, composition and function of the brain, it remains the least understood organ of the human body. To gain a deeper comprehension of the neural system scientists aim to simplistically reconstruct the tissue by assembling it in vitro from basic building blocks using a tissue engineering approach. Our group developed a tissue-engineered silk and collagen-based 3D brain-like model resembling the white and gray matter of the cortex. The model consists of silk porous sponge, which is pre-seeded with rat brain-derived neurons, immersed in soft collagen matrix. Polarized neuronal outgrowth and network formation is observed with separate axonal and cell body localization. This compartmental architecture allows for the unique development of niches mimicking native neural tissue, thus enabling research on neuronal network assembly, axonal guidance, cell-cell and cell-matrix interactions and electrical functions.
Collapse
Affiliation(s)
| | - Disha Sood
- Department of Biomedical Engineering, Tufts University
| | | | - James D White
- Department of Biomedical Engineering, Tufts University
| | - Min Tang-Schomer
- Department of Pediatrics, University of Connecticut Health Center & Connecticut Children's Medical Center
| | | |
Collapse
|
36
|
Le Goff GC, Lee J, Gupta A, Hill WA, Doyle PS. High-Throughput Contact Flow Lithography. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2015; 2:1500149. [PMID: 27980910 PMCID: PMC5115321 DOI: 10.1002/advs.201500149] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 05/19/2015] [Indexed: 05/20/2023]
Abstract
High-throughput fabrication of graphically encoded hydrogel microparticles is achieved by combining flow contact lithography in a multichannel microfluidic device and a high capacity 25 mm LED UV source. Production rates of chemically homogeneous particles are improved by two orders of magnitude. Additionally, the custom-built contact lithography instrument provides an affordable solution for patterning complex microstructures on surfaces.
Collapse
Affiliation(s)
- Gaelle C Le Goff
- Department of Chemical Engineering Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA; Novartis Institutes for Biomedical Research 250 Massachusetts Avenue Cambridge MA 02139 USA
| | - Jiseok Lee
- Department of Chemical Engineering Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA; School of Energy and Chemical Engineering Ulsan National Institute of Science and Technology Eonyang-eup Ulju-gun Ulsan 689-798 South Korea
| | - Ankur Gupta
- Department of Chemical Engineering Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| | - William Adam Hill
- Novartis Institutes for Biomedical Research 250 Massachusetts Avenue Cambridge MA 02139 USA
| | - Patrick S Doyle
- Department of Chemical Engineering Massachusetts Institute of Technology 77 Massachusetts Avenue Cambridge MA 02139 USA
| |
Collapse
|
37
|
Chen P, Güven S, Usta OB, Yarmush ML, Demirci U. Biotunable acoustic node assembly of organoids. Adv Healthc Mater 2015; 4:1937-43. [PMID: 26149464 PMCID: PMC4731612 DOI: 10.1002/adhm.201500279] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/21/2015] [Indexed: 11/06/2022]
Abstract
Bioengineering of 3D microtissues from cell spheroids is demonstrated by employing the vibration of acoustic standing waves and its hydrodynamic effect at the bottom of a liquid-carrier chamber. A large number of cell spheroids (>10(4) ) are assembled in seconds into a closely packed structure in a scaffold-free fashion under nodal pattern of the standing waves in a fluidic environment.
Collapse
Affiliation(s)
- Pu Chen
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304
| | - Sinan Güven
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304
| | - Osman Berk Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114
| | - Martin L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ 08854
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304
| |
Collapse
|
38
|
Li B, Wang L, Xu F, Gang X, Demirci U, Wei D, Li Y, Feng Y, Jia D, Zhou Y. Hydrosoluble, UV-crosslinkable and injectable chitosan for patterned cell-laden microgel and rapid transdermal curing hydrogel in vivo. Acta Biomater 2015; 22:59-69. [PMID: 25917845 DOI: 10.1016/j.actbio.2015.04.026] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 04/16/2015] [Accepted: 04/19/2015] [Indexed: 01/05/2023]
Abstract
Natural and biodegradable chitosan with unique amino groups has found widespread applications in tissue engineering and drug delivery. However, its applications have been limited by the poor solubility of native chitosan in neutral pH solution, which subsequently fails to achieve cell-laden hydrogel at physiological pH. To address this, we incorporated UV crosslinking ability in chitosan, allowing fabrication of patterned cell-laden and rapid transdermal curing hydrogel in vivo. The hydrosoluble, UV crosslinkable and injectable N-methacryloyl chitosan (N-MAC) was synthesized via single-step chemoselective N-acylation reaction, which simultaneously endowed chitosan with well solubility in neutral pH solution, UV crosslinkable ability and injectability. The solubility of N-MAC in neutral pH solution increased 2.21-fold with substitution degree increasing from 10.9% to 28.4%. The N-MAC allowed fabrication of cell-laden microgels with on-demand patterns via photolithography, and the cell viability in N-MAC hydrogel maintained 96.3 ± 1.3% N-MAC allowed rapid transdermal curing hydrogel in vivo within 60s through minimally invasive clinical surgery. Histological analysis revealed that low-dose UV irradiation hardly induced skin injury and acute inflammatory response disappeared after 7 days. N-MAC would allow rapid, robust and cost-effective fabrication of patterned cell-laden polysaccharide microgels with unique amino groups serving as building blocks for tissue engineering and rapid transdermal curing hydrogel in vivo for localized and sustained protein delivery.
Collapse
|
39
|
Wang L, Li Y, Chen B, Liu S, Li M, Zheng L, Wang P, Lu TJ, Xu F. Patterning Cellular Alignment through Stretching Hydrogels with Programmable Strain Gradients. ACS APPLIED MATERIALS & INTERFACES 2015; 7:15088-15097. [PMID: 26079936 DOI: 10.1021/acsami.5b04450] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The graded mechanical properties (e.g., stiffness and stress/strain) of excellular matrix play an important role in guiding cellular alignment, as vital in tissue reconstruction with proper functions. Though various methods have been developed to engineer a graded mechanical environment to study its effect on cellular behaviors, most of them failed to distinguish stiffness effect from stress/strain effect during mechanical loading. Here, we construct a mechanical environment with programmable strain gradients by using a hydrogel of a linear elastic property. When seeding cells on such hydrogels, we demonstrate that the pattern of cellular alignment can be rather precisely tailored by substrate strains. The experiment is in consistency with a theoritical prediction when assuming that focal adhesions (FAs) would drive a cell to reorient to the directions where they are most stable. A fundamental theory has also been developed and is excellent in agreement with the complete temporal alignment of cells. This work not only provides important insights into the cellular response to the local mechanical microenvironment but can also be utilized to engineer patterned cellular alignment that can be critical in tissue remodeling and regenerative medicine applications.
Collapse
Affiliation(s)
| | | | - Bin Chen
- ∥Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, People's Republic of China
| | | | | | | | - Pengfei Wang
- §Qian Xuesen Laboratory of Space Technology, China Academy of Space Technology, Beijing 100094, People's Republic of China
| | | | | |
Collapse
|
40
|
Guven S, Chen P, Inci F, Tasoglu S, Erkmen B, Demirci U. Multiscale assembly for tissue engineering and regenerative medicine. Trends Biotechnol 2015; 33:269-279. [PMID: 25796488 DOI: 10.1016/j.tibtech.2015.02.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/30/2015] [Accepted: 02/03/2015] [Indexed: 01/11/2023]
Abstract
Our understanding of cell biology and its integration with materials science has led to technological innovations in the bioengineering of tissue-mimicking grafts that can be utilized in clinical and pharmaceutical applications. Bioengineering of native-like multiscale building blocks provides refined control over the cellular microenvironment, thus enabling functional tissues. In this review, we focus on assembling building blocks from the biomolecular level to the millimeter scale. We also provide an overview of techniques for assembling molecules, cells, spheroids, and microgels and achieving bottom-up tissue engineering. Additionally, we discuss driving mechanisms for self- and guided assembly to create micro-to-macro scale tissue structures.
Collapse
Affiliation(s)
- Sinan Guven
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Pu Chen
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Fatih Inci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Savas Tasoglu
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
| | - Burcu Erkmen
- BAMM Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford School of Medicine, Palo Alto, CA 94304, USA
- BAMM Laboratory, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
41
|
Wang L, Li Y, Huang G, Zhang X, Pingguan-Murphy B, Gao B, Lu TJ, Xu F. Hydrogel-based methods for engineering cellular microenvironment with spatiotemporal gradients. Crit Rev Biotechnol 2015; 36:553-65. [PMID: 25641330 DOI: 10.3109/07388551.2014.993588] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Natural cellular microenvironment consists of spatiotemporal gradients of multiple physical (e.g. extracellular matrix stiffness, porosity and stress/strain) and chemical cues (e.g. morphogens), which play important roles in regulating cell behaviors including spreading, proliferation, migration, differentiation and apoptosis, especially for pathological processes such as tumor formation and progression. Therefore, it is essential to engineer cellular gradient microenvironment incorporating various gradients for the fabrication of normal and pathological tissue models in vitro. In this article, we firstly review the development of engineering cellular physical and chemical gradients with cytocompatible hydrogels in both two-dimension and three-dimension formats. We then present current advances in the application of engineered gradient microenvironments for the fabrication of disease models in vitro. Finally, concluding remarks and future perspectives for engineering cellular gradients are given.
Collapse
Affiliation(s)
- Lin Wang
- a MOE Key Laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Yuhui Li
- a MOE Key Laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Guoyou Huang
- a MOE Key Laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Xiaohui Zhang
- a MOE Key Laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Belinda Pingguan-Murphy
- c Department of Biomedical Engineering , Faculty of Engineering, University of Malaya , Kuala Lumpur , Malaysia , and
| | - Bin Gao
- a MOE Key Laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China .,d Department of Endocrinology and Metabolism , Xijing Hospital, Fourth Military Medical University , Xi'an , China
| | - Tian Jian Lu
- b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| | - Feng Xu
- a MOE Key Laboratory of Biomedical Information Engineering , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , China .,b Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University , Xi'an , China
| |
Collapse
|
42
|
Schmidt D, Cho YK. Natural photoreceptors and their application to synthetic biology. Trends Biotechnol 2015; 33:80-91. [DOI: 10.1016/j.tibtech.2014.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/19/2014] [Accepted: 10/20/2014] [Indexed: 01/22/2023]
|
43
|
Abstract
The brain remains one of the most important but least understood tissues in our body, in part because of its complexity as well as the limitations associated with in vivo studies. Although simpler tissues have yielded to the emerging tools for in vitro 3D tissue cultures, functional brain-like tissues have not. We report the construction of complex functional 3D brain-like cortical tissue, maintained for months in vitro, formed from primary cortical neurons in modular 3D compartmentalized architectures with electrophysiological function. We show that, on injury, this brain-like tissue responds in vitro with biochemical and electrophysiological outcomes that mimic observations in vivo. This modular 3D brain-like tissue is capable of real-time nondestructive assessments, offering previously unidentified directions for studies of brain homeostasis and injury.
Collapse
|
44
|
Gurkan UA, El Assal R, Yildiz SE, Sung Y, Trachtenberg AJ, Kuo WP, Demirci U. Engineering anisotropic biomimetic fibrocartilage microenvironment by bioprinting mesenchymal stem cells in nanoliter gel droplets. Mol Pharm 2014; 11:2151-9. [PMID: 24495169 PMCID: PMC4096228 DOI: 10.1021/mp400573g] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past decade, bioprinting has emerged as a promising patterning strategy to organize cells and extracellular components both in two and three dimensions (2D and 3D) to engineer functional tissue mimicking constructs. So far, tissue printing has neither been used for 3D patterning of mesenchymal stem cells (MSCs) in multiphase growth factor embedded 3D hydrogels nor been investigated phenotypically in terms of simultaneous differentiation into different cell types within the same micropatterned 3D tissue constructs. Accordingly, we demonstrated a biochemical gradient by bioprinting nanoliter droplets encapsulating human MSCs, bone morphogenetic protein 2 (BMP-2), and transforming growth factor β1 (TGF- β1), engineering an anisotropic biomimetic fibrocartilage microenvironment. Assessment of the model tissue construct displayed multiphasic anisotropy of the incorporated biochemical factors after patterning. Quantitative real time polymerase chain reaction (qRT-PCR) results suggested genomic expression patterns leading to simultaneous differentiation of MSC populations into osteogenic and chondrogenic phenotype within the multiphasic construct, evidenced by upregulation of osteogenesis and condrogenesis related genes during in vitro culture. Comprehensive phenotypic network and pathway analysis results, which were based on genomic expression data, indicated activation of differentiation related mechanisms, via signaling pathways, including TGF, BMP, and vascular endothelial growth factor.
Collapse
Affiliation(s)
- Umut A Gurkan
- Case Biomanufacturing and Microfabrication Laboratory, Mechanical and Aerospace Engineering Department, Department of Orthopaedics, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | | | | | | | | | | | | |
Collapse
|
45
|
Zhao R, Chen CS, Reich DH. Force-driven evolution of mesoscale structure in engineered 3D microtissues and the modulation of tissue stiffening. Biomaterials 2014; 35:5056-64. [PMID: 24630092 DOI: 10.1016/j.biomaterials.2014.02.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/12/2014] [Indexed: 12/16/2022]
Abstract
The complex structures of tissues determine their mechanical strength. In engineered tissues formed through self-assembly in a mold, artificially imposed boundary constraints have been found to induce anisotropic clustering of the cells and the extracellular matrix in local regions. To understand how such tissue remodeling at the intermediate length-scale (mesoscale) affects tissue stiffening, we used a novel microtissue mechanical testing system to manipulate the remodeling of the tissue structures and to measure the subsequent changes in tissue stiffness. Microtissues were formed through cell driven self-assembly of collagen matrix in arrays of micro-patterned wells, each containing two flexible micropillars that measured the microtissues' contractile forces and elastic moduli via magnetic actuation. We manipulated tissue remodeling by inducing myofibroblast differentiation with TGF-β1, by varying the micropillar spring constants or by blocking cell contractility with blebbistatin and collagen cross-linking with BAPN. We showed that increased anisotropic compaction of the collagen matrix, caused by increased micropillar spring constant or elevated cell contraction force, contributed to tissue stiffening. Conversely, collagen matrix and tissue stiffness were not affected by inhibition of cell-generated contraction forces. Together, these measurements showed that mesoscale tissue remodeling is an important middle step linking tissue compaction forces and tissue stiffening.
Collapse
Affiliation(s)
- Ruogang Zhao
- Department of Physics and Astronomy, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| | - Christopher S Chen
- Department of Bioengineering, University of Pennsylvania, 510 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA 19104, USA
| | - Daniel H Reich
- Department of Physics and Astronomy, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
46
|
Han YL, Wang S, Zhang X, Li Y, Huang G, Qi H, Pingguan-Murphy B, Li Y, Lu TJ, Xu F. Engineering physical microenvironment for stem cell based regenerative medicine. Drug Discov Today 2014; 19:763-73. [PMID: 24508818 DOI: 10.1016/j.drudis.2014.01.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/20/2014] [Accepted: 01/27/2014] [Indexed: 12/13/2022]
Abstract
Regenerative medicine has rapidly evolved over the past decade owing to its potential applications to improve human health. Targeted differentiations of stem cells promise to regenerate a variety of tissues and/or organs despite significant challenges. Recent studies have demonstrated the vital role of the physical microenvironment in regulating stem cell fate and improving differentiation efficiency. In this review, we summarize the main physical cues that are crucial for controlling stem cell differentiation. Recent advances in the technologies for the construction of physical microenvironment and their implications in controlling stem cell fate are also highlighted.
Collapse
Affiliation(s)
- Yu Long Han
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Shuqi Wang
- Brigham Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaohui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Hao Qi
- Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and training Center, Beijing, 100094, China
| | - Tian Jian Lu
- Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China.
| |
Collapse
|
47
|
Tasoglu S, Diller E, Guven S, Sitti M, Demirci U. Untethered micro-robotic coding of three-dimensional material composition. Nat Commun 2014; 5:3124. [PMID: 24469115 PMCID: PMC3947548 DOI: 10.1038/ncomms4124] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 12/16/2013] [Indexed: 11/09/2022] Open
Abstract
Complex functional materials with three-dimensional micro- or nano-scale dynamic compositional features are prevalent in nature. However, the generation of three-dimensional functional materials composed of both soft and rigid microstructures, each programmed by shape and composition, is still an unsolved challenge. Here we describe a method to code complex materials in three-dimensions with tunable structural, morphological and chemical features using an untethered magnetic micro-robot remotely controlled by magnetic fields. This strategy allows the micro-robot to be introduced to arbitrary microfluidic environments for remote two- and three-dimensional manipulation. We demonstrate the coding of soft hydrogels, rigid copper bars, polystyrene beads and silicon chiplets into three-dimensional heterogeneous structures. We also use coded microstructures for bottom-up tissue engineering by generating cell-encapsulating constructs.
Collapse
Affiliation(s)
- S. Tasoglu
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - E. Diller
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - S. Guven
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - M. Sitti
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - U. Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
48
|
Unal M, Alapan Y, Jia H, Varga AG, Angelino K, Aslan M, Sayin I, Han C, Jiang Y, Zhang Z, Gurkan UA. Micro and Nano-Scale Technologies for Cell Mechanics. Nanobiomedicine (Rij) 2014; 1:5. [PMID: 30023016 PMCID: PMC6029242 DOI: 10.5772/59379] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/18/2014] [Indexed: 01/09/2023] Open
Abstract
Cell mechanics is a multidisciplinary field that bridges cell biology, fundamental mechanics, and micro and nanotechnology, which synergize to help us better understand the intricacies and the complex nature of cells in their native environment. With recent advances in nanotechnology, microfabrication methods and micro-electro-mechanical-systems (MEMS), we are now well situated to tap into the complex micro world of cells. The field that brings biology and MEMS together is known as Biological MEMS (BioMEMS). BioMEMS take advantage of systematic design and fabrication methods to create platforms that allow us to study cells like never before. These new technologies have been rapidly advancing the study of cell mechanics. This review article provides a succinct overview of cell mechanics and comprehensively surveys micro and nano-scale technologies that have been specifically developed for and are relevant to the mechanics of cells. Here we focus on micro and nano-scale technologies, and their applications in biology and medicine, including imaging, single cell analysis, cancer cell mechanics, organ-on-a-chip systems, pathogen detection, implantable devices, neuroscience and neurophysiology. We also provide a perspective on the future directions and challenges of technologies that relate to the mechanics of cells.
Collapse
Affiliation(s)
- Mustafa Unal
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, USA
| | - Yunus Alapan
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, USA
- Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Cleveland, USA
| | - Hao Jia
- Department of Biology, Case Western Reserve University, Cleveland, USA
| | - Adrienn G. Varga
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Keith Angelino
- Department of Civil Engineering, Case Western Reserve University, Cleveland, USA
| | - Mahmut Aslan
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, USA
- Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Cleveland, USA
| | - Ismail Sayin
- Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Cleveland, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Chanjuan Han
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, USA
| | - Yanxia Jiang
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, USA
| | - Zhehao Zhang
- Department of Civil Engineering, Case Western Reserve University, Cleveland, USA
| | - Umut A. Gurkan
- Department of Electrical Engineering and Computer Science, Case Western Reserve University, Cleveland, USA
- Case Biomanufacturing and Microfabrication Laboratory, Case Western Reserve University, Cleveland, USA
- Department of Orthopaedics, Case Western Reserve University, Cleveland, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, USA
| |
Collapse
|
49
|
Liu W, Li Y, Wang T, Li D, Fang L, Zhu S, Shen H, Zhang J, Sun H, Yang B. Elliptical polymer brush ring array mediated protein patterning and cell adhesion on patterned protein surfaces. ACS APPLIED MATERIALS & INTERFACES 2013; 5:12587-12593. [PMID: 24256492 DOI: 10.1021/am403808s] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
This paper presents a novel method to fabricate elliptical ring arrays of proteins. The protein arrays are prepared by covalently grafting proteins to the polymer brush ring arrays which are prepared by the techniques combining colloidal lithography dewetting and surface initiated atom-transfer radical polymerization (SI-ATRP). Through this method, the parameters of protein patterns, such as height, wall thickness, periods, and distances between two elliptical rings, can be finely regulated. In addition, the sample which contains the elliptical protein ring arrays can be prepared over a large area up to 1 cm(2), and the protein on the ring maintains its biological activity. The as-prepared ring and elliptical ring arrays (ERAs) of fibronectin can promote cell adhesion and may have an active effect on the formation of the actin cytoskeleton.
Collapse
Affiliation(s)
- Wendong Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University , Changchun 130012, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kolewe ME, Park H, Gray C, Ye X, Langer R, Freed LE. 3D structural patterns in scalable, elastomeric scaffolds guide engineered tissue architecture. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:4459-65. [PMID: 23765688 PMCID: PMC3954574 DOI: 10.1002/adma.201301016] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/24/2013] [Indexed: 05/22/2023]
Abstract
Microfabricated elastomeric scaffolds with 3D structural patterns are created by semiautomated layer-by-layer assembly of planar polymer sheets with through-pores. The mesoscale interconnected pore architectures governed by the relative alignment of layers are shown to direct cell and muscle-like fiber orientation in both skeletal and cardiac muscle, enabling scale up of tissue constructs towards clinically relevant dimensions.
Collapse
Affiliation(s)
- Martin E. Kolewe
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hyoungshin Park
- Microsystems Development and Microfabrication Process Engineering Groups, Charles Stark Draper Laboratory, Cambridge, MA 02139, USA
| | - Caprice Gray
- Microsystems Development and Microfabrication Process Engineering Groups, Charles Stark Draper Laboratory, Cambridge, MA 02139, USA
| | - Xiaofeng Ye
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert Langer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lisa E. Freed
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Microsystems Development and Microfabrication Process Engineering Groups, Charles Stark Draper Laboratory, Cambridge, MA 02139, USA
| |
Collapse
|