101
|
Bruggeman KF, Moriarty N, Dowd E, Nisbet DR, Parish CL. Harnessing stem cells and biomaterials to promote neural repair. Br J Pharmacol 2019; 176:355-368. [PMID: 30444942 PMCID: PMC6329623 DOI: 10.1111/bph.14545] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/16/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023] Open
Abstract
With the limited capacity for self-repair in the adult CNS, efforts to stimulate quiescent stem cell populations within discrete brain regions, as well as harness the potential of stem cell transplants, offer significant hope for neural repair. These new cells are capable of providing trophic cues to support residual host populations and/or replace those cells lost to the primary insult. However, issues with low-level adult neurogenesis, cell survival, directed differentiation and inadequate reinnervation of host tissue have impeded the full potential of these therapeutic approaches and their clinical advancement. Biomaterials offer novel approaches to stimulate endogenous neurogenesis, as well as for the delivery and support of neural progenitor transplants, providing a tissue-appropriate physical and trophic milieu for the newly integrating cells. In this review, we will discuss the various approaches by which bioengineered scaffolds may improve stem cell-based therapies for repair of the CNS.
Collapse
Affiliation(s)
- K F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of EngineeringThe Australian National UniversityCanberraACTAustralia
| | - N Moriarty
- Pharmacology and Therapeutics and Galway Neuroscience CentreNational University of Ireland GalwayGalwayIreland
| | - E Dowd
- Pharmacology and Therapeutics and Galway Neuroscience CentreNational University of Ireland GalwayGalwayIreland
| | - D R Nisbet
- Laboratory of Advanced Biomaterials, Research School of EngineeringThe Australian National UniversityCanberraACTAustralia
| | - C L Parish
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVICAustralia
| |
Collapse
|
102
|
Subbiah R, Guldberg RE. Materials Science and Design Principles of Growth Factor Delivery Systems in Tissue Engineering and Regenerative Medicine. Adv Healthc Mater 2019; 8:e1801000. [PMID: 30398700 DOI: 10.1002/adhm.201801000] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/13/2018] [Indexed: 01/22/2023]
Abstract
Growth factors (GFs) are signaling molecules that direct cell development by providing biochemical cues for stem cell proliferation, migration, and differentiation. GFs play a key role in tissue regeneration, but one major limitation of GF-based therapies is dosage-related adverse effects. Additionally, the clinical applications and efficacy of GFs are significantly affected by the efficiency of delivery systems and other pharmacokinetic factors. Hence, it is crucial to design delivery systems that provide optimal activity, stability, and tunable delivery for GFs. Understanding the physicochemical properties of the GFs and the biomaterials utilized for the development of biomimetic GF delivery systems is critical for GF-based regeneration. Many different delivery systems have been developed to achieve tunable delivery kinetics for single or multiple GFs. The identification of ideal biomaterials with tunable properties for spatiotemporal delivery of GFs is still challenging. This review characterizes the types, properties, and functions of GFs, the materials science of widely used biomaterials, and various GF loading strategies to comprehensively summarize the current delivery systems for tunable spatiotemporal delivery of GFs aimed for tissue regeneration applications. This review concludes by discussing fundamental design principles for GF delivery vehicles based on the interactive physicochemical properties of the proteins and biomaterials.
Collapse
Affiliation(s)
- Ramesh Subbiah
- Parker H. Petit Institute for Bioengineering and Bioscience; George W. Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience; George W. Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
- Phil and Penny Knight Campus for Accelerating Scientific Impact; 6231 University of Oregon; Eugene OR 97403 USA
| |
Collapse
|
103
|
Oksdath M, Perrin SL, Bardy C, Hilder EF, DeForest CA, Arrua RD, Gomez GA. Review: Synthetic scaffolds to control the biochemical, mechanical, and geometrical environment of stem cell-derived brain organoids. APL Bioeng 2018; 2:041501. [PMID: 31069322 PMCID: PMC6481728 DOI: 10.1063/1.5045124] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/31/2018] [Indexed: 01/16/2023] Open
Abstract
Stem cell-derived brain organoids provide a powerful platform for systematic studies of tissue functional architecture and the development of personalized therapies. Here, we review key advances at the interface of soft matter and stem cell biology on synthetic alternatives to extracellular matrices. We emphasize recent biomaterial-based strategies that have been proven advantageous towards optimizing organoid growth and controlling the geometrical, biomechanical, and biochemical properties of the organoid's three-dimensional environment. We highlight systems that have the potential to increase the translational value of region-specific brain organoid models suitable for different types of manipulations and high-throughput applications.
Collapse
Affiliation(s)
- Mariana Oksdath
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | - Sally L. Perrin
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| | | | - Emily F. Hilder
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Cole A. DeForest
- Department of Chemical Engineering and Department of Bioengineering, University of Washington, Seattle, Washington 98195-1750, USA
| | - R. Dario Arrua
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia
| | - Guillermo A. Gomez
- Centre for Cancer Biology, South Australia Pathology and University of South Australia, Adelaide 5001, Australia
| |
Collapse
|
104
|
Park MH, Subbiah R, Kwon MJ, Kim WJ, Kim SH, Park K, Lee K. The three dimensional cues-integrated-biomaterial potentiates differentiation of human mesenchymal stem cells. Carbohydr Polym 2018; 202:488-496. [DOI: 10.1016/j.carbpol.2018.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/29/2018] [Accepted: 09/05/2018] [Indexed: 12/20/2022]
|
105
|
Correia CR, Reis RL, Mano JF. Design Principles and Multifunctionality in Cell Encapsulation Systems for Tissue Regeneration. Adv Healthc Mater 2018; 7:e1701444. [PMID: 30102458 DOI: 10.1002/adhm.201701444] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Cell encapsulation systems are being increasingly applied as multifunctional strategies to regenerate tissues. Lessons afforded with encapsulation systems aiming to treat endocrine diseases seem to be highly valuable for the tissue engineering and regenerative medicine (TERM) systems of today, in which tissue regeneration and biomaterial integration are key components. Innumerous multifunctional systems for cell compartmentalization are being proposed to meet the specific needs required in the TERM field. Herein is reviewed the variable geometries proposed to produce cell encapsulation strategies toward tissue regeneration, including spherical and fiber-shaped systems, and other complex shapes and arrangements that better mimic the highly hierarchical organization of native tissues. The application of such principles in the TERM field brings new possibilities for the development of highly complex systems, which holds tremendous promise for tissue regeneration. The complex systems aim to recreate adequate environmental signals found in native tissue (in particular during the regenerative process) to control the cellular outcome, and conferring multifunctional properties, namely the incorporation of bioactive molecules and the ability to create smart and adaptative systems in response to different stimuli. The new multifunctional properties of such systems that are being employed to fulfill the requirements of the TERM field are also discussed.
Collapse
Affiliation(s)
- Clara R. Correia
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - João F. Mano
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| |
Collapse
|
106
|
Lu JY, Zhu QY, Zhang XX, Zhang FR, Huang WT, Ding XZ, Xia LQ, Luo HQ, Li NB. Directly repurposing waste optical discs with prefabricated nanogrooves as a platform for investigation of cell-substrate interactions and guiding neuronal growth. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 160:273-281. [PMID: 29852430 DOI: 10.1016/j.ecoenv.2018.05.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/08/2018] [Accepted: 05/24/2018] [Indexed: 06/08/2023]
Abstract
Due to rapid change in information technology, many consumer electronics become electronic waste which is the fastest-growing pollution problems worldwide. In fact, many discarded electronics with prefabricated micro/nanostructures may provide a good basis to fulfill special needs of other fields, such as tissue engineering, biosensors, and energy. Herein, to take waste optical discs as an example, we demonstrate that discarded electronics can be directly repurposed as highly anisotropic platforms for in vitro investigation of cell behaviors, such as cell adhesion, cell alignment, and cell-cell interactions. The PC12 cells cultured on biocompatible DVD polycarbonate layers with flat and grooved morphology show a distinct cell morphology, indicating the topographical cue of nanogrooves plays a key role in guidance of neurites growth. By further monitoring cell morphology and alignment of PC12 cells cultured on the DVD nanogrooves at different differentiation times, we find that cell contact interaction with nanotopographies is dynamically adjustable with differentiation time from initial disorder to final order. This study adds a new dimension to not only solving the problems of supply of materials and fabrication of nanopatterns in neural tissue engineering, but may also offering a new promising way of waste minimization or reuse for environmental protection.
Collapse
Affiliation(s)
- Jiao Yang Lu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Qiu Yan Zhu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Xin Xing Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Fu Rui Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Wei Tao Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China.
| | - Xue Zhi Ding
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Li Qiu Xia
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, PR China
| | - Hong Qun Luo
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region (Ministry of Education), School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Nian Bing Li
- Key Laboratory of Eco-environments in Three Gorges Reservoir Region (Ministry of Education), School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| |
Collapse
|
107
|
Wang X, Liu C. Fibrin Hydrogels for Endothelialized Liver Tissue Engineering with a Predesigned Vascular Network. Polymers (Basel) 2018; 10:E1048. [PMID: 30960973 PMCID: PMC6403613 DOI: 10.3390/polym10101048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/04/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022] Open
Abstract
The design and manufacture of a branched vascular network is essential for bioartificial organ implantation, which provides nutrients and removes metabolites for multi-cellular tissues. In the present study, we present a technology to manufacture endothelialized liver tissues using a fibrin hydrogel and a rotational combined mold. Both hepatocytes and adipose-derived stem cells (ADSCs) encapsulated in a fibrin hydrogel were assembled into a spindle construct with a predesigned multi-branched vascular network. An external overcoat of poly(dl-lactic-co-glycolic acid) was used to increase the mechanical properties of the construct as well as to act as an impervious and isolating membrane around the construct. Cell survivability reached 100% in the construct after 6 days of in vitro culture. ADSCs in the spindle construct were engaged into endothelial cells/tissues using a cocktail growth factor engagement approach. Mechanical property comparison and permeability evaluation tests all indicated that this was a viable complex organ containing more than two heterogeneous tissue types and a functional vascular network. It is, therefore, the first time an implantable bioartificial liver, i.e., endothelialized liver tissue, along with a hierarchical vascular network, has been created.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China.
- Center of Organ Manufacturing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China.
| | - Chang Liu
- Tianjin Mifang Science & Technology Ltd., Wuqing 301701, China.
| |
Collapse
|
108
|
|
109
|
Truong VX, Li F, Forsythe JS. Visible Light Activation of Nucleophilic Thiol-X Addition via Thioether Bimane Photocleavage for Polymer Cross-Linking. Biomacromolecules 2018; 19:4277-4285. [DOI: 10.1021/acs.biomac.8b01153] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Vinh X. Truong
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Fanyi Li
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
- CSIRO Manufacturing, Bayview Avenue, Clayton, Victoria 3168, Australia
| | - John S. Forsythe
- Department of Materials Science and Engineering, Monash Institute of Medical Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
110
|
Visible light controls cell adhesion on a photoswitchable biointerface. Colloids Surf B Biointerfaces 2018; 169:41-48. [DOI: 10.1016/j.colsurfb.2018.04.062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/08/2018] [Accepted: 04/30/2018] [Indexed: 01/02/2023]
|
111
|
Yamamoto S, Ikegami H, Yamaguchi K, Nakanishi J. A Dynamic Biomaterial Based on a 2-Nitrobenzyl Derivative with a tert
-Butyl Substituent at the Benzyl Position: Rapid Response and Minimized Phototoxicity. CHEMPHOTOCHEM 2018. [DOI: 10.1002/cptc.201800087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Shota Yamamoto
- International Center for Materials Nanoarchitectonics (WPI-MANA); National Institute for Materials Science (NIMS); 1-1 Namiki, Tsukuba Ibaraki 305-0044 Japan
| | - Hiroki Ikegami
- Department of Chemistry; Kanagawa University; 2946 Tsuchiya, Hiratsuka Kanagawa 259-1293 Japan
| | - Kazuo Yamaguchi
- Department of Chemistry; Kanagawa University; 2946 Tsuchiya, Hiratsuka Kanagawa 259-1293 Japan
| | - Jun Nakanishi
- International Center for Materials Nanoarchitectonics (WPI-MANA); National Institute for Materials Science (NIMS); 1-1 Namiki, Tsukuba Ibaraki 305-0044 Japan
- Graduate School of Advanced Science and Engineering; Waseda University; 3-4-1 Okubo, Shinjuku-ku Tokyo 169-8555 Japan
| |
Collapse
|
112
|
Jensen G, Morrill C, Huang Y. 3D tissue engineering, an emerging technique for pharmaceutical research. Acta Pharm Sin B 2018; 8:756-766. [PMID: 30258764 PMCID: PMC6148716 DOI: 10.1016/j.apsb.2018.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering and the tissue engineering model have shown promise in improving methods of drug delivery, drug action, and drug discovery in pharmaceutical research for the attenuation of the central nervous system inflammatory response. Such inflammation contributes to the lack of regenerative ability of neural cells, as well as the temporary and permanent loss of function associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and traumatic brain injury. This review is focused specifically on the recent advances in the tissue engineering model made by altering scaffold biophysical and biochemical properties for use in the treatment of neurodegenerative diseases. A portion of this article will also be spent on the review of recent progress made in extracellular matrix decellularization as a new and innovative scaffold for disease treatment.
Collapse
Affiliation(s)
| | | | - Yu Huang
- Department of Biological Engineering, Utah State University, Logan, UT, 84322, USA
| |
Collapse
|
113
|
Abstract
The conjugation of biomolecules can impart materials with the bioactivity necessary to modulate specific cell behaviors. While the biological roles of particular polypeptide, oligonucleotide, and glycan structures have been extensively reviewed, along with the influence of attachment on material structure and function, the key role played by the conjugation strategy in determining activity is often overlooked. In this review, we focus on the chemistry of biomolecule conjugation and provide a comprehensive overview of the key strategies for achieving controlled biomaterial functionalization. No universal method exists to provide optimal attachment, and here we will discuss both the relative advantages and disadvantages of each technique. In doing so, we highlight the importance of carefully considering the impact and suitability of a particular technique during biomaterial design.
Collapse
Affiliation(s)
- Christopher D. Spicer
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden
| | - E. Thomas Pashuck
- NJ
Centre for Biomaterials, Rutgers University, 145 Bevier Road, Piscataway, New Jersey United States
| | - Molly M. Stevens
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden
- Department
of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London, United Kingdom
| |
Collapse
|
114
|
Jivan F, Fabela N, Davis Z, Alge DL. Orthogonal click reactions enable the synthesis of ECM-mimetic PEG hydrogels without multi-arm precursors. J Mater Chem B 2018; 6:4929-4936. [PMID: 30746148 PMCID: PMC6368189 DOI: 10.1039/c8tb01399c] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Click chemistry reactions have become an important tool for synthesizing user-defined hydrogels consisting of poly(ethylene glycol) (PEG) and bioactive peptides for tissue engineering. However, because click crosslinking proceeds via a step-growth mechanism, multi-arm telechelic precursors are required, which has some disadvantages. Here, we report for the first time that this requirement can be circumvented to create PEG-peptide hydrogels solely from linear precursors through the use of two orthogonal click reactions, the thiol-maleimide Michael addition and thiol-norbornene click reaction. The rapid kinetics of both click reactions allowed for quick formation of norbornene-functionalized PEG-peptide block copolymers via Michael addition, which were subsequently photocrosslinked into hydrogels with a dithiol linker. Characterization and in vitro testing demonstrated that the hydrogels have highly tunable physicochemical properties and excellent cytocompatiiblity. In addition, stoichiometric control over the crosslinking reaction can be leveraged to leave unreacted norbornene groups in the hydrogel for subsequent hydrogel functionalization via bioorthogonal inverse-electron demand Diels-Alder click reactions with s-tetrazines. After selectively capping norbornene groups in a user-defined region with cysteine, this feature was leveraged for protein patterning. Collectively, these results demonstrate that our novel chemical strategy is a simple and versatile approach to the development of hydrogels for tissue engineering that could be useful for a variety of applications.
Collapse
Affiliation(s)
- Faraz Jivan
- Texas A&M University, Department of Biomedical Engineering, 5045 Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, United States
| | - Natalia Fabela
- Texas A&M University, Department of Biomedical Engineering, 5045 Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, United States
| | - Zachary Davis
- North Carolina State University, Department of Materials Science and Engineering, 911 Partners Way, Raleigh, NC 27606, United States
| | - Daniel L Alge
- Texas A&M University, Department of Biomedical Engineering, 5045 Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, United States
- Texas A&M University, Department of Materials Science and Engineering, 3003 TAMU, College Station, TX, 77843, United States
| |
Collapse
|
115
|
Borowiec J, Hampl J, Singh S, Haefner S, Friedel K, Mai P, Brauer D, Ruther F, Liverani L, Boccaccini AR, Schober A. 3D Microcontact Printing for Combined Chemical and Topographical Patterning on Porous Cell Culture Membrane. ACS APPLIED MATERIALS & INTERFACES 2018; 10:22857-22865. [PMID: 29883090 DOI: 10.1021/acsami.8b06585] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Micrometer-scale biochemical or topographical patterning is commonly used to guide the cell attachment and growth, but the ability to combine these patterns into an integrated surface with defined chemical and geometrical characteristics still remains a technical challenge. Here, we present a technical solution for simultaneous construction of 3D morphologies, in the form of channels, on porous membranes along with precise transfer of extracellular matrix proteins into the channels to create patterns with geometrically restricting features. By combining the advantages of microthermoforming and microcontact printing, this technique offers a unique patterning process that provides spatiotemporal control over morphological and chemical feature in a single step. By use of our 3D-microcontact printing (3DμCP), determined microstructures like channels with different depths and widths even with more complex patterns can be fabricated. Collagen, fibronectin, and laminin were successfully transferred inside the predesigned geometries, and the validity of the process was confirmed by antibody staining. Cells cultivated on 3DμCP patterned polycarbonate membrane have shown selective adhesion and growth. This technique offers a novel tool for creating freeform combinatorial patterning on the thermoformable surface.
Collapse
Affiliation(s)
- Justyna Borowiec
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Joerg Hampl
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Sukhdeep Singh
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Sebastian Haefner
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Karin Friedel
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Patrick Mai
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Dana Brauer
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| | - Florian Ruther
- Institute of Biomaterials, Department of Materials Science and Engineering , University of Erlangen-Nuremberg , 91058 Erlangen , Germany
| | - Liliana Liverani
- Institute of Biomaterials, Department of Materials Science and Engineering , University of Erlangen-Nuremberg , 91058 Erlangen , Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering , University of Erlangen-Nuremberg , 91058 Erlangen , Germany
| | - Andreas Schober
- Department of Nanobiosystem Technology, Institute of Micro- and Nanotechnologies MacroNano, Institute of Chemistry and Biotechnology , Ilmenau University of Technology , 98693 Ilmenau , Germany
| |
Collapse
|
116
|
Ma X, Liu J, Zhu W, Tang M, Lawrence N, Yu C, Gou M, Chen S. 3D bioprinting of functional tissue models for personalized drug screening and in vitro disease modeling. Adv Drug Deliv Rev 2018; 132:235-251. [PMID: 29935988 PMCID: PMC6226327 DOI: 10.1016/j.addr.2018.06.011] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 05/04/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023]
Abstract
3D bioprinting is emerging as a promising technology for fabricating complex tissue constructs with tailored biological components and mechanical properties. Recent advances have enabled scientists to precisely position materials and cells to build functional tissue models for in vitro drug screening and disease modeling. This review presents state-of-the-art 3D bioprinting techniques and discusses the choice of cell source and biomaterials for building functional tissue models that can be used for personalized drug screening and disease modeling. In particular, we focus on 3D-bioprinted liver models, cardiac tissues, vascularized constructs, and cancer models for their promising applications in medical research, drug discovery, toxicology, and other pre-clinical studies.
Collapse
Affiliation(s)
- Xuanyi Ma
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Justin Liu
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Wei Zhu
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Min Tang
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Natalie Lawrence
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Claire Yu
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, PR China
| | - Shaochen Chen
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of NanoEngineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, PR China.
| |
Collapse
|
117
|
Blache U, Ehrbar M. Inspired by Nature: Hydrogels as Versatile Tools for Vascular Engineering. Adv Wound Care (New Rochelle) 2018; 7:232-246. [PMID: 29984113 PMCID: PMC6032659 DOI: 10.1089/wound.2017.0760] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/21/2022] Open
Abstract
Significance: Diseases related to vascular malfunction, hyper-vascularization, or lack of vascularization are among the leading causes of morbidity and mortality. Engineered, vascularized tissues as well as angiogenic growth factor-releasing hydrogels could replace defective tissues. Further, treatments and testing of novel vascular therapeutics will benefit significantly from models that allow for the study of vascularized tissues under physiological relevant in vitro conditions. Recent Advances: Inspired by fibrin, the provisional matrix during wound healing, naturally derived and synthetic hydrogel scaffolds have been developed for vascular engineering. Today, engineers and biologists use commercially available hydrogels to pre-vascularize tissues, to control the delivery of angiogenic growth factors, and to establish vascular diseases models. Critical Issue: For clinical translation, pre-vascularized tissue constructs must be sufficiently large and stable to substitute function-relevant tissue defects and integrate with host vascular perfusion. Moreover, the continuous integration of knowhow from basic vascular biology with innovative, tailorable materials and advanced manufacturing technologies is key to achieving near-physiological tissue models and new treatments to control vascularization. Future Directions: For transplantation, engineered tissues must comprise hierarchically organized vascular trees of different caliber and function. The development of novel vascularization-promoting or -inhibiting therapeutics will benefit from physiologically relevant vessel models. In addition, tissue models representing treatment-relevant vascular tissue functions will increase the capacity to screen for therapeutic compounds and will significantly reduce the need for animals for their validation.
Collapse
Affiliation(s)
- Ulrich Blache
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
118
|
Curtis C, Toghani D, Wong B, Nance E. Colloidal stability as a determinant of nanoparticle behavior in the brain. Colloids Surf B Biointerfaces 2018; 170:673-682. [PMID: 29986264 DOI: 10.1016/j.colsurfb.2018.06.050] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 05/17/2018] [Accepted: 06/22/2018] [Indexed: 01/07/2023]
Abstract
Drug delivery to the brain is challenging due to a highly regulated blood-brain barrier (BBB) and a complex brain microenvironment. Nanoparticles, due to their tailorability, provide promising platforms to enhance therapeutic delivery and achieve controlled release and disease-specific localization in the brain. However, we have yet to fully understand the complex interactions between nanoparticles and the biological environments in which they operate. It is important to perform a systematic study to characterize nanoparticle behavior as a function of ion composition, concentration, and pH in cerebrospinal fluid (CSF). These could alter nanoparticle biological identity and influence diffusive capability and cellular uptake. In this study, poly(ethylene glycol) (PEG)-coated and carboxyl-coated polystyrene (PS-PEG and PS-COOH respectively) nanoparticles (NPs) were used to evaluate the aggregation kinetics, colloidal stability, and diffusive capability of nanoparticles in conditions relevant to the brain microenvironment. Size, surface charge, and surface coating were varied in a range of CSF ion concentrations and compositions, pH conditions, and temperatures. Small changes in calcium concentration and pH destabilize nanoparticles in CSF. However, PS-PEG NPs remain stable over a wider variety of conditions than PS-COOH NPs, and have higher diffusion capabilities in both agarose gels, an in vitro model of the brain microenvironment, and an organotypic brain tissue slice model. These results demonstrate the need for steric stabilization to maintain nanoparticle colloidal stability in a wide range of conditions. Importantly, colloidal stabilization allows for increased diffusive capability and can be used to predict diffusive behavior in the brain microenvironment.
Collapse
Affiliation(s)
- Chad Curtis
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, United States
| | - Dorsa Toghani
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, United States
| | - Ben Wong
- Math Academy, College of Engineering, University of Washington, Seattle, WA, 98195, United States
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, United States.
| |
Collapse
|
119
|
Dicker KT, Song J, Moore AC, Zhang H, Li Y, Burris DL, Jia X, Fox JM. Core-shell patterning of synthetic hydrogels via interfacial bioorthogonal chemistry for spatial control of stem cell behavior. Chem Sci 2018; 9:5394-5404. [PMID: 30009011 PMCID: PMC6009435 DOI: 10.1039/c8sc00495a] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/24/2018] [Indexed: 12/29/2022] Open
Abstract
A new technique is described for the patterning of cell-guidance cues in synthetic extracellular matrices (ECM) for tissue engineering applications. Using s-tetrazine modified hyaluronic acid (HA), bis-trans-cyclooctene (TCO) crosslinkers and monofunctional TCO conjugates, interfacial bioorthogonal crosslinking was used to covalently functionalize hydrogels as they were synthesized at the liquid-gel interface. Through temporally controlled introduction of TCO conjugates during the crosslinking process, the enzymatic degradability, cell adhesivity, and mechanical properties of the synthetic microenvironment can be tuned with spatial precision. Using human mesenchymal stem cells (hMSCs) and hydrogels with a core-shell structure, we demonstrated the ability of the synthetic ECM with spatially defined guidance cues to modulate cell morphology in a biomimetic fashion. This new method for the spatially resolved introduction of cell-guidance cues for the establishment of functional tissue constructs complements existing methods that require UV-light or specialized equipment.
Collapse
Affiliation(s)
- K T Dicker
- Department of Materials Science and Engineering , University of Delaware , DuPont Hall , Newark , DE 19716 , USA . ;
| | - J Song
- Department of Materials Science and Engineering , University of Delaware , DuPont Hall , Newark , DE 19716 , USA . ;
| | - A C Moore
- Department of Biomedical Engineering , University of Delaware , Colburn Lab , Newark , DE 19716 , USA
| | - H Zhang
- Department of Chemistry and Biochemistry , University of Delaware , Brown Lab , Newark , DE 19716 , USA
| | - Y Li
- Department of Chemistry and Biochemistry , University of Delaware , Brown Lab , Newark , DE 19716 , USA
| | - D L Burris
- Department of Biomedical Engineering , University of Delaware , Colburn Lab , Newark , DE 19716 , USA
- Department of Mechanical Engineering , University of Delaware , Spencer Lab , Newark , DE 19716 , USA
| | - X Jia
- Department of Materials Science and Engineering , University of Delaware , DuPont Hall , Newark , DE 19716 , USA . ;
- Department of Biomedical Engineering , University of Delaware , Colburn Lab , Newark , DE 19716 , USA
| | - J M Fox
- Department of Materials Science and Engineering , University of Delaware , DuPont Hall , Newark , DE 19716 , USA . ;
- Department of Chemistry and Biochemistry , University of Delaware , Brown Lab , Newark , DE 19716 , USA
| |
Collapse
|
120
|
Lau HK, Paul A, Sidhu I, Li L, Sabanayagam CR, Parekh SH, Kiick KL. Microstructured Elastomer-PEG Hydrogels via Kinetic Capture of Aqueous Liquid-Liquid Phase Separation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1701010. [PMID: 29938180 PMCID: PMC6010786 DOI: 10.1002/advs.201701010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/22/2018] [Indexed: 05/31/2023]
Abstract
Heterogeneous hydrogels with desired matrix complexity are studied for a variety of biomimetic materials. Despite the range of such microstructured materials described, few methods permit independent control over microstructure and microscale mechanics by precisely controlled, single-step processing methods. Here, a phototriggered crosslinking methodology that traps microstructures in liquid-liquid phase-separated solutions of a highly elastomeric resilin-like polypeptide (RLP) and poly(ethylene glycol) (PEG) is reported. RLP-rich domains of various diameters can be trapped in a PEG continuous phase, with the kinetics of domain maturation dependent on the degree of acrylation. The chemical composition of both hydrogel phases over time is assessed via in situ hyperspectral coherent Raman microscopy, with equilibrium concentrations consistent with the compositions derived from NMR-measured coexistence curves. Atomic force microscopy reveals that the local mechanical properties of the two phases evolve over time, even as the bulk modulus of the material remains constant, showing that the strategy permits control of mechanical properties on micrometer length scales, of relevance in generating mechanically robust materials for a range of applications. As one example, the successful encapsulation, localization, and survival of primary cells are demonstrated and suggest the potential application of phase-separated RLP-PEG hydrogels in regenerative medicine applications.
Collapse
Affiliation(s)
- Hang Kuen Lau
- Department of Materials Science and EngineeringUniversity of Delaware201 DuPont HallNewarkDE19716USA
| | - Alexandra Paul
- Department of Biology and Biological EngineeringChalmers University of TechnologyGothenburgSE‐412 96Sweden
- Department of Molecular SpectroscopyMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Ishnoor Sidhu
- Department of Biological SciencesUniversity of DelawareNewarkDE19716USA
| | - Linqing Li
- Department of Materials Science and EngineeringUniversity of Delaware201 DuPont HallNewarkDE19716USA
| | | | - Sapun H. Parekh
- Department of Molecular SpectroscopyMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Kristi L. Kiick
- Department of Materials Science and EngineeringUniversity of Delaware201 DuPont HallNewarkDE19716USA
- Delaware Biotechnology Institute15 Innovation WayNewarkDE19711USA
| |
Collapse
|
121
|
Jeon O, Lee K, Alsberg E. Spatial Micropatterning of Growth Factors in 3D Hydrogels for Location-Specific Regulation of Cellular Behaviors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800579. [PMID: 29782703 PMCID: PMC6238642 DOI: 10.1002/smll.201800579] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/29/2018] [Indexed: 05/26/2023]
Abstract
Growth factors are potent stimuli for regulating cell function in tissue engineering strategies, but spatially patterning their presentation in 3D in a facile manner using a single material is challenging. Micropatterning is an attractive tool to modulate the cellular microenvironment with various biochemical and physical cues and study their effects on stem cell behaviors. Implementing heparin's ability to immobilize growth factors, dual-crosslinkable alginate hydrogels are micropatterned in 3D with photocrosslinkable heparin substrates with various geometries and micropattern sizes, and their capability to establish 3D micropatterns of growth factors within the hydrogels is confirmed. This 3D micropatterning method could be applied to various heparin binding growth factors, such as fibroblast growth factor-2, vascular endothelial growth factor, transforming growth factor-betas and bone morphogenetic proteins while retaining the hydrogel's natural degradability and cytocompability. Stem cells encapsulated within these micropatterned hydrogels have exhibited spatially localized growth and differentiation responses corresponding to various growth factor patterns, demonstrating the versatility of the approach in controlling stem cell behavior for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Keewon Lee
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
122
|
Li J, Weber E, Guth-Gundel S, Schuleit M, Kuttler A, Halleux C, Accart N, Doelemeyer A, Basler A, Tigani B, Wuersch K, Fornaro M, Kneissel M, Stafford A, Freedman BR, Mooney DJ. Tough Composite Hydrogels with High Loading and Local Release of Biological Drugs. Adv Healthc Mater 2018; 7:e1701393. [PMID: 29441702 PMCID: PMC6192424 DOI: 10.1002/adhm.201701393] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/09/2018] [Indexed: 11/10/2022]
Abstract
Hydrogels are under active development for controlled drug delivery, but their clinical translation is limited by low drug loading capacity, deficiencies in mechanical toughness and storage stability, and poor control over the drug release that often results in burst release and short release duration. This work reports a design of composite clay hydrogels, which simultaneously achieve a spectrum of mechanical, storage, and drug loading/releasing properties to address the critical needs from translational perspectives. The clay nanoparticles provide large surface areas to adsorb biological drugs, and assemble into microparticles that are physically trapped within and toughen hydrogel networks. The composite hydrogels demonstrate feasibility of storage, and extended release of large quantities of an insulin-like growth factor-1 mimetic protein (8 mg mL-1 ) over four weeks. The release rate is primarily governed by ionic exchange and can be upregulated by low pH, which is typical for injured tissues. A rodent model of Achilles tendon injury is used to demonstrate that the composite hydrogels allow for highly extended and localized release of biological drugs in vivo, while demonstrating biodegradation and biocompatibility. These attributes make the composite hydrogel a promising system for drug delivery and regenerative medicine.
Collapse
Affiliation(s)
- Jianyu Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Department of Mechanical Engineering, McGill University, Montreal, QC, H3A 0C3, Canada
| | - Eckhard Weber
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Sabine Guth-Gundel
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Michael Schuleit
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Andreas Kuttler
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Christine Halleux
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Nathalie Accart
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Arno Doelemeyer
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Anne Basler
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Bruno Tigani
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Kuno Wuersch
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Mara Fornaro
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Michaela Kneissel
- Novartis Institutes for BioMedical Research, Fabrikstrasse 2, Novartis Campus, Basel, CH, 4056, Switzerland
| | - Alexander Stafford
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
123
|
Hickey K, Stabenfeldt SE. Using biomaterials to modulate chemotactic signaling for central nervous system repair. Biomed Mater 2018; 13:044106. [PMID: 29411713 PMCID: PMC5991092 DOI: 10.1088/1748-605x/aaad82] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemotaxis enables cellular communication and movement within the body. This review focuses on exploiting chemotaxis as a tool for repair of the central nervous system (CNS) damaged from injury and/or degenerative diseases. Chemokines and factors alone may initiate repair following CNS injury/disease, but exogenous administration may enhance repair and promote regeneration. This review will discuss critical chemotactic molecules and factors that may promote neural regeneration. Additionally, this review highlights how biomaterials can impact the presentation and delivery of chemokines and growth factors to alter the regenerative response.
Collapse
Affiliation(s)
- Kassondra Hickey
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| | | |
Collapse
|
124
|
Lambert CR, Nijsure D, Huynh V, Wylie RG. Hydrogels with reversible chemical environments for
in vitro
cell culture. Biomed Mater 2018; 13:045002. [DOI: 10.1088/1748-605x/aab45d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
125
|
Qin XH, Wang X, Rottmar M, Nelson BJ, Maniura-Weber K. Near-Infrared Light-Sensitive Polyvinyl Alcohol Hydrogel Photoresist for Spatiotemporal Control of Cell-Instructive 3D Microenvironments. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1705564. [PMID: 29333748 DOI: 10.1002/adma.201705564] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/22/2017] [Indexed: 06/07/2023]
Abstract
Advanced hydrogel systems that allow precise control of cells and their 3D microenvironments are needed in tissue engineering, disease modeling, and drug screening. Multiphoton lithography (MPL) allows true 3D microfabrication of complex objects, but its biological application requires a cell-compatible hydrogel resist that is sufficiently photosensitive, cell-degradable, and permissive to support 3D cell growth. Here, an extremely photosensitive cell-responsive hydrogel composed of peptide-crosslinked polyvinyl alcohol (PVA) is designed to expand the biological applications of MPL. PVA hydrogels are formed rapidly by ultraviolet light within 1 min in the presence of cells, providing fully synthetic matrices that are instructive for cell-matrix remodeling, multicellular morphogenesis, and protease-mediated cell invasion. By focusing a multiphoton laser into a cell-laden PVA hydrogel, cell-instructive extracellular cues are site-specifically attached to the PVA matrix. Cell invasion is thus precisely guided in 3D with micrometer-scale spatial resolution. This robust hydrogel enables, for the first time, ultrafast MPL of cell-responsive synthetic matrices at writing speeds up to 50 mm s-1 . This approach should enable facile photochemical construction and manipulation of 3D cellular microenvironments with unprecedented flexibility and precision.
Collapse
Affiliation(s)
- Xiao-Hua Qin
- Laboratory for Biointerfaces, Empa-Swiss Federal Laboratories for Materials Science and Technology, CH-9014, St. Gallen, Switzerland
| | - Xiaopu Wang
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zürich, CH-8092, Zürich, Switzerland
| | - Markus Rottmar
- Laboratory for Biointerfaces, Empa-Swiss Federal Laboratories for Materials Science and Technology, CH-9014, St. Gallen, Switzerland
| | - Bradley J Nelson
- Multi-Scale Robotics Lab, Institute of Robotics and Intelligent Systems, ETH Zürich, CH-8092, Zürich, Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa-Swiss Federal Laboratories for Materials Science and Technology, CH-9014, St. Gallen, Switzerland
| |
Collapse
|
126
|
Fisher SA, Tam RY, Fokina A, Mahmoodi MM, Distefano MD, Shoichet MS. Photo-immobilized EGF chemical gradients differentially impact breast cancer cell invasion and drug response in defined 3D hydrogels. Biomaterials 2018; 178:751-766. [PMID: 29452913 DOI: 10.1016/j.biomaterials.2018.01.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/15/2017] [Accepted: 01/20/2018] [Indexed: 01/08/2023]
Abstract
Breast cancer cell invasion is influenced by growth factor concentration gradients in the tumor microenvironment. However, studying the influence of growth factor gradients on breast cancer cell invasion is challenging due to both the complexities of in vivo models and the difficulties in recapitulating the tumor microenvironment with defined gradients using in vitro models. A defined hyaluronic acid (HA)-based hydrogel crosslinked with matrix metalloproteinase (MMP) cleavable peptides and modified with multiphoton labile nitrodibenzofuran (NDBF) was synthesized to photochemically immobilize epidermal growth factor (EGF) gradients. We demonstrate that EGF gradients can differentially influence breast cancer cell invasion and drug response in cell lines with different EGF receptor (EGFR) expression levels. Photopatterned EGF gradients increase the invasion of moderate EGFR expressing MDA-MB-231 cells, reduce invasion of high EGFR expressing MDA-MB-468 cells, and have no effect on invasion of low EGFR-expressing MCF-7 cells. We evaluate MDA-MB-231 and MDA-MB-468 cell response to the clinically tested EGFR inhibitor, cetuximab. Interestingly, the cellular response to cetuximab is completely different on the EGF gradient hydrogels: cetuximab decreases MDA-MB-231 cell invasion but increases MDA-MB-468 cell invasion and cell number, thus demonstrating the importance of including cell-microenvironment interactions when evaluating drug targets.
Collapse
Affiliation(s)
- Stephanie A Fisher
- The Donnelly Centre for Cellular and Biomolecular Research, Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, 160 College Street, Toronto Ontario, M5S 3E1, Canada
| | - Roger Y Tam
- The Donnelly Centre for Cellular and Biomolecular Research, Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, 160 College Street, Toronto Ontario, M5S 3E1, Canada
| | - Ana Fokina
- The Donnelly Centre for Cellular and Biomolecular Research, Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, 160 College Street, Toronto Ontario, M5S 3E1, Canada
| | - M Mohsen Mahmoodi
- Department of Chemistry, University of Minnesota, Minneapolis MN, 55455, USA
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis MN, 55455, USA
| | - Molly S Shoichet
- The Donnelly Centre for Cellular and Biomolecular Research, Department of Chemical Engineering and Applied Chemistry, Institute of Biomaterials and Biomedical Engineering, University of Toronto, 160 College Street, Toronto Ontario, M5S 3E1, Canada.
| |
Collapse
|
127
|
Bruggeman KF, Williams RJ, Nisbet DR. Dynamic and Responsive Growth Factor Delivery from Electrospun and Hydrogel Tissue Engineering Materials. Adv Healthc Mater 2018; 7. [PMID: 29193871 DOI: 10.1002/adhm.201700836] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/04/2017] [Indexed: 12/21/2022]
Abstract
Tissue engineering scaffolds are designed to mimic physical, chemical, and biological features of the extracellular matrix, thereby providing a constant support that is crucial to improved regenerative medicine outcomes. Beyond mechanical and structural support, the next generation of these materials must also consider the more dynamic presentation and delivery of drugs or growth factors to guide new and regenerating tissue development. These two aspects are explored expansively separately, but they must interact synergistically to achieve optimal regeneration. This review explores common tissue engineering materials types, electrospun polymers and hydrogels, and strategies used for incorporating drug delivery systems into these scaffolds.
Collapse
Affiliation(s)
- Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials; Research School of Engineering; The Australian National University; Canberra ACT 2601 Australia
| | - Richard J. Williams
- School of Engineering; RMIT University; Melbourne VIC 3001 Australia
- Biofab3D; Aikenhead Center for Medical Discovery; St. Vincent's Hospital; Melbourne VIC 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials; Research School of Engineering; The Australian National University; Canberra ACT 2601 Australia
- Biofab3D; Aikenhead Center for Medical Discovery; St. Vincent's Hospital; Melbourne VIC 3065 Australia
| |
Collapse
|
128
|
Thomas D, O'Brien T, Pandit A. Toward Customized Extracellular Niche Engineering: Progress in Cell-Entrapment Technologies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1703948. [PMID: 29194781 DOI: 10.1002/adma.201703948] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/12/2017] [Indexed: 06/07/2023]
Abstract
The primary aim in tissue engineering is to repair, replace, and regenerate dysfunctional tissues to restore homeostasis. Cell delivery for repair and regeneration is gaining impetus with our understanding of constructing tissue-like environments. However, the perpetual challenge is to identify innovative materials or re-engineer natural materials to model cell-specific tissue-like 3D modules, which can seamlessly integrate and restore functions of the target organ. To devise an optimal functional microenvironment, it is essential to define how simple is complex enough to trigger tissue regeneration or restore cellular function. Here, the purposeful transition of cell immobilization from a cytoprotection point of view to that of a cell-instructive approach is examined, with advances in the understanding of cell-material interactions in a 3D context, and with a view to further application of the knowledge for the development of newer and complex hierarchical tissue assemblies for better examination of cell behavior and offering customized cell-based therapies for tissue engineering.
Collapse
Affiliation(s)
- Dilip Thomas
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
129
|
Li W, Yan Z, Ren J, Qu X. Manipulating cell fate: dynamic control of cell behaviors on functional platforms. Chem Soc Rev 2018; 47:8639-8684. [DOI: 10.1039/c8cs00053k] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the recent advances and new horizons in the dynamic control of cell behaviors on functional platforms and their applications.
Collapse
Affiliation(s)
- Wen Li
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Science
- Changchun
- P. R. China
| | - Zhengqing Yan
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Science
- Changchun
- P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Science
- Changchun
- P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization
- Changchun Institute of Applied Chemistry
- Chinese Academy of Science
- Changchun
- P. R. China
| |
Collapse
|
130
|
Yao H, Wang J, Mi S. Photo Processing for Biomedical Hydrogels Design and Functionality: A Review. Polymers (Basel) 2017; 10:E11. [PMID: 30966045 PMCID: PMC6415176 DOI: 10.3390/polym10010011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 01/20/2023] Open
Abstract
A large number of opportunities for biomedical hydrogel design and functionality through photo-processing have stretched the limits of innovation. As both photochemical understanding and engineering technologies continue to develop, more complicated geometries and spatiotemporal manipulations can be realized through photo-exposure, producing multifunctional hydrogels with specific chemical, biological and physical characteristics for the achievement of biomedical goals. This report describes the role that light has recently played in the synthesis and functionalization of biomedical hydrogels and primarily the design of photoresponsive hydrogels via different chemical reactions (photo crosslinking and photo degradation) and conventional light curing processes (micropatterning, stereolithography and two/multiphoton techniques) as well as typical biomedical applications of the hydrogels (cell culture, differentiation and in vivo vascularization) and their promising future.
Collapse
Affiliation(s)
- Hongyi Yao
- Biomanufacturing Engineering Laboratory, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.
| | - Jieqiong Wang
- Biomanufacturing Engineering Laboratory, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.
| | - Shengli Mi
- Biomanufacturing Engineering Laboratory, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.
- Open FIESTA Center, Tsinghua University, Shenzhen 518055, China.
| |
Collapse
|
131
|
Macroscopic Supramolecular Assembly and Its Applications. CHINESE JOURNAL OF POLYMER SCIENCE 2017. [DOI: 10.1007/s10118-018-2069-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
132
|
Hu JL, Todhunter ME, LaBarge MA, Gartner ZJ. Opportunities for organoids as new models of aging. J Cell Biol 2017; 217:39-50. [PMID: 29263081 PMCID: PMC5748992 DOI: 10.1083/jcb.201709054] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/13/2017] [Accepted: 11/27/2017] [Indexed: 01/02/2023] Open
Abstract
The biology of aging is challenging to study, particularly in humans. As a result, model organisms are used to approximate the physiological context of aging in humans. However, the best model organisms remain expensive and time-consuming to use. More importantly, they may not reflect directly on the process of aging in people. Human cell culture provides an alternative, but many functional signs of aging occur at the level of tissues rather than cells and are therefore not readily apparent in traditional cell culture models. Organoids have the potential to effectively balance between the strengths and weaknesses of traditional models of aging. They have sufficient complexity to capture relevant signs of aging at the molecular, cellular, and tissue levels, while presenting an experimentally tractable alternative to animal studies. Organoid systems have been developed to model many human tissues and diseases. Here we provide a perspective on the potential for organoids to serve as models for aging and describe how current organoid techniques could be applied to aging research.
Collapse
Affiliation(s)
- Jennifer L Hu
- University of California Berkeley-University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA
| | - Michael E Todhunter
- Center for Cancer and Aging, Beckman Research Institute at City of Hope, Duarte, CA
| | - Mark A LaBarge
- Center for Cancer and Aging, Beckman Research Institute at City of Hope, Duarte, CA
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA .,National Science Foundation Center for Cellular Construction, University of California San Francisco, San Francisco, CA.,Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
133
|
Abstract
Hydrogels mimic many of the physical properties of soft tissue and are widely used biomaterials for tissue engineering and regenerative medicine. Synthetic hydrogels have been developed to recapitulate many of the healthy and diseased states of native tissues and can be used as a cell scaffold to study the effect of matricellular interactions in vitro. However, these matrices often fail to capture the dynamic and heterogenous nature of the in vivo environment, which varies spatially and during events such as development and disease. To address this deficiency, a variety of manufacturing and processing techniques are being adapted to the biomaterials setting. Among these, photochemistry is particularly well suited because these reactions can be performed in precise three-dimensional space and at specific moments in time. This spatiotemporal control over chemical reactions can also be performed over a range of cell- and tissue-relevant length scales with reactions that proceed efficiently and harmlessly at ambient conditions. This review will focus on the use of photochemical reactions to create dynamic hydrogel environments, and how these dynamic environments are being used to investigate and direct cell behavior.
Collapse
Affiliation(s)
- Tobin E Brown
- Department of Chemical and Biological Engineering, University of Colorado Boulder, USA.
| | | |
Collapse
|
134
|
Ning R, Zhuang Q, Lin JM. Biomaterial-Based Microfluidics for Cell Culture and Analysis. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-981-10-5394-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
135
|
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: 479] [Impact Index Per Article: 68.4] [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
|
136
|
Valentin TM, Leggett SE, Chen PY, Sodhi JK, Stephens LH, McClintock HD, Sim JY, Wong IY. Stereolithographic printing of ionically-crosslinked alginate hydrogels for degradable biomaterials and microfluidics. LAB ON A CHIP 2017; 17:3474-3488. [PMID: 28906525 PMCID: PMC5636682 DOI: 10.1039/c7lc00694b] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
3D printed biomaterials with spatial and temporal functionality could enable interfacial manipulation of fluid flows and motile cells. However, such dynamic biomaterials are challenging to implement since they must be responsive to multiple, biocompatible stimuli. Here, we show stereolithographic printing of hydrogels using noncovalent (ionic) crosslinking, which enables reversible patterning with controlled degradation. We demonstrate this approach using sodium alginate, photoacid generators and various combinations of divalent cation salts, which can be used to tune the hydrogel degradation kinetics, pattern fidelity, and mechanical properties. This approach is first utilized to template perfusable microfluidic channels within a second encapsulating hydrogel for T-junction and gradient devices. The presence and degradation of printed alginate microstructures were further verified to have minimal toxicity on epithelial cells. Degradable alginate barriers were used to direct collective cell migration from different initial geometries, revealing differences in front speed and leader cell formation. Overall, this demonstration of light-based 3D printing using non-covalent crosslinking may enable adaptive and stimuli-responsive biomaterials, which could be utilized for bio-inspired sensing, actuation, drug delivery, and tissue engineering.
Collapse
Affiliation(s)
- Thomas M Valentin
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular & Nanoscale Innovation, Brown University, 184 Hope St, Box D, Providence, RI 02912, USA.
| | | | | | | | | | | | | | | |
Collapse
|
137
|
Dorsey TB, Grath A, Xu C, Hong Y, Dai G. Patterning Bioactive Proteins or Peptides on Hydrogel Using Photochemistry for Biological Applications. J Vis Exp 2017. [PMID: 28994764 DOI: 10.3791/55873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
There are many biological stimuli that can influence cell behavior and stem cell differentiation. General cell culture approaches rely on soluble factors within the medium to control cell behavior. However, soluble additions cannot mimic certain signaling motifs, such as matrix-bound growth factors, cell-cell signaling, and spatial biochemical cues, which are common influences on cells. Furthermore, biophysical properties of the matrix, such as substrate stiffness, play important roles in cell fate, which is not easily manipulated using conventional cell culturing practices. In this method, we describe a straightforward protocol to provide patterned bioactive proteins on synthetic polyethylene glycol (PEG) hydrogels using photochemistry. This platform allows for the independent control of substrate stiffness and spatial biochemical cues. These hydrogels can achieve a large range of physiologically relevant stiffness values. Additionally, the surfaces of these hydrogels can be photopatterned with bioactive peptides or proteins via thiol-ene click chemistry reactions. These methods have been optimized to retain protein function after surface immobilization. This is a versatile protocol that can be applied to any protein or peptide of interest to create a variety of patterns. Finally, cells seeded onto the surfaces of these bioactive hydrogels can be monitored over time as they respond to spatially specific signals.
Collapse
Affiliation(s)
- Taylor B Dorsey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Department of Bioengineering, Northeastern University
| | - Alexander Grath
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute
| | - Cancan Xu
- Department of Bioengineering, University of Texas at Arlington
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute; Department of Bioengineering, Northeastern University;
| |
Collapse
|
138
|
Liu Y, Ma Y, Zhang J, Xie Q, Wang Z, Yu S, Yuan Y, Liu C. MBG-Modified β-TCP Scaffold Promotes Mesenchymal Stem Cells Adhesion and Osteogenic Differentiation via a FAK/MAPK Signaling Pathway. ACS APPLIED MATERIALS & INTERFACES 2017; 9:30283-30296. [PMID: 28820575 DOI: 10.1021/acsami.7b02466] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The β-TCP scaffold has been widely used as a bone graft substitute, but the traditional PMMA molding method-induced undesirable mechanical strength and poor interconnectivity still have not been addressed until now. In this study, a MBG-based PU foam templating method was developed to fabricate β-TCP scaffolds with desirable microtopography. The MBG gel, as both binder and modifier, prepared by a modified sol-gel method with controlled viscosity is incorporated with β-TCP powder and thereafter is impregnated into PU foam. The resultant hybrid scaffolds exhibited interconnected macropores (200-500 μm) and distinctive micropores (0.2-1.5 μm), especially for the TCP/25MBG (with 25 wt % content MBG). As expected, the compression strength of β-TCP/MBG composite scaffolds was enhanced with increasing MBG content, and TCP/50MBG (with 50 wt % content MBG) exhibited almost 100-fold enhancement compared to the pure β-TCP. Intriguingly, the cell affinity and osteogenic capacity of rBMSCs were also dramatically improved the best on TCP/25MBG. Further investigation found that the subtle, grainy-like microtopography, not the chemical composition, of the TCP/25MBG favored the adsorption of Fn and expression of integrin α5β1 and further facilitated FA formation and the expression of p-FAK, following activation of the MAPK/ERK signaling pathway and ultimately upregulated expression of osteogenic genes. Further in vivo experiments confirmed the promoted osteogenesis of TCP/25MBG in vivo. The results suggest that such a novel MBG-based PU foam templating method offers new guidance to construct hierarchically porous scaffolds, and the prepared MBG-modified β-TCP scaffold will have great potential for future use in bone tissue regeneration.
Collapse
Affiliation(s)
| | | | | | - Qing Xie
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200011, People's Republic of China
| | - Zi Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai 200011, People's Republic of China
| | | | | | | |
Collapse
|
139
|
Intestinal Stem Cell Niche Insights Gathered from Both In Vivo and Novel In Vitro Models. Stem Cells Int 2017; 2017:8387297. [PMID: 29081810 PMCID: PMC5610807 DOI: 10.1155/2017/8387297] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022] Open
Abstract
Intestinal stem cells are located at the base of the crypts and are surrounded by a complex structure called niche. This environment is composed mainly of epithelial cells and stroma which provides signals that govern cell maintenance, proliferation, and differentiation. Understanding how the niche regulates stem cell fate by controlling developmental signaling pathways will help us to define how stem cells choose between self-renewal and differentiation and how they maintain their undifferentiated state. Tractable in vitro assay systems, which reflect the complexity of the in vivo situation but provide higher level of control, would likely be crucial in identifying new players and mechanisms controlling stem cell function. Knowledge of the intestinal stem cell niche gathered from both in vivo and novel in vitro models may help us improve therapies for tumorigenesis and intestinal damage and make autologous intestinal transplants a feasible clinical practice.
Collapse
|
140
|
Leijten J, Seo J, Yue K, Santiago GTD, Tamayol A, Ruiz-Esparza GU, Shin SR, Sharifi R, Noshadi I, Álvarez MM, Zhang YS, Khademhosseini A. Spatially and Temporally Controlled Hydrogels for Tissue Engineering. MATERIALS SCIENCE & ENGINEERING. R, REPORTS : A REVIEW JOURNAL 2017; 119:1-35. [PMID: 29200661 PMCID: PMC5708586 DOI: 10.1016/j.mser.2017.07.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Recent years have seen tremendous advances in the field of hydrogel-based biomaterials. One of the most prominent revolutions in this field has been the integration of elements or techniques that enable spatial and temporal control over hydrogels' properties and functions. Here, we critically review the emerging progress of spatiotemporal control over biomaterial properties towards the development of functional engineered tissue constructs. Specifically, we will highlight the main advances in the spatial control of biomaterials, such as surface modification, microfabrication, photo-patterning, and three-dimensional (3D) bioprinting, as well as advances in the temporal control of biomaterials, such as controlled release of molecules, photocleaving of proteins, and controlled hydrogel degradation. We believe that the development and integration of these techniques will drive the engineering of next-generation engineered tissues.
Collapse
Affiliation(s)
- Jeroen Leijten
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Jungmok Seo
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Kan Yue
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Grissel Trujillo-de Santiago
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Microsystems Technologies Laboratories, MIT, Cambridge, 02139, MA, USA
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, CP 64849, Monterrey, Nuevo León, México
| | - Ali Tamayol
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Guillermo U. Ruiz-Esparza
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Roholah Sharifi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Iman Noshadi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Mario Moisés Álvarez
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Microsystems Technologies Laboratories, MIT, Cambridge, 02139, MA, USA
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey at Monterrey, CP 64849, Monterrey, Nuevo León, México
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
141
|
Zhang YS, Zhu C, Xia Y. Inverse Opal Scaffolds and Their Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:10.1002/adma.201701115. [PMID: 28649794 PMCID: PMC5581229 DOI: 10.1002/adma.201701115] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 03/23/2017] [Indexed: 05/04/2023]
Abstract
Three-dimensional porous scaffolds play a pivotal role in tissue engineering and regenerative medicine by functioning as biomimetic substrates to manipulate cellular behaviors. While many techniques have been developed to fabricate porous scaffolds, most of them rely on stochastic processes that typically result in scaffolds with pores uncontrolled in terms of size, structure, and interconnectivity, greatly limiting their use in tissue regeneration. Inverse opal scaffolds, in contrast, possess uniform pores inheriting from the template comprised of a closely packed lattice of monodispersed microspheres. The key parameters of such scaffolds, including architecture, pore structure, porosity, and interconnectivity, can all be made uniform across the same sample and among different samples. In conjunction with a tight control over pore sizes, inverse opal scaffolds have found widespread use in biomedical applications. In this review, we provide a detailed discussion on this new class of advanced materials. After a brief introduction to their history and fabrication, we highlight the unique advantages of inverse opal scaffolds over their non-uniform counterparts. We then showcase their broad applications in tissue engineering and regenerative medicine, followed by a summary and perspective on future directions.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Chunlei Zhu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
- School of Chemistry and Biochemistry, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
142
|
Tsurkan MV, Jungnickel C, Schlierf M, Werner C. Forbidden Chemistry: Two-Photon Pathway in [2+2] Cycloaddition of Maleimides. J Am Chem Soc 2017; 139:10184-10187. [DOI: 10.1021/jacs.7b04484] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mikhail V. Tsurkan
- Max
Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research, Hohe Strasse 6, 01069 Dresden, Germany
| | - Christiane Jungnickel
- Max
Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research, Hohe Strasse 6, 01069 Dresden, Germany
- B CUBE
- Center for Molecular Bioengineering, Technische Universität Dresden, Arnoldstraße 18, 01307 Dresden, Germany
| | - Michael Schlierf
- B CUBE
- Center for Molecular Bioengineering, Technische Universität Dresden, Arnoldstraße 18, 01307 Dresden, Germany
| | - Carsten Werner
- Max
Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research, Hohe Strasse 6, 01069 Dresden, Germany
- Center
for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- B CUBE
- Center for Molecular Bioengineering, Technische Universität Dresden, Arnoldstraße 18, 01307 Dresden, Germany
| |
Collapse
|
143
|
Guo C, Kim H, Ovadia EM, Mourafetis CM, Yang M, Chen W, Kloxin AM. Bio-orthogonal conjugation and enzymatically triggered release of proteins within multi-layered hydrogels. Acta Biomater 2017; 56:80-90. [PMID: 28391052 PMCID: PMC5510749 DOI: 10.1016/j.actbio.2017.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 02/02/2023]
Abstract
Hydrogels are facile architectures for the controlled presentation of proteins with far-reaching applications, from fundamental biological studies in three-dimensional culture to new regenerative medicine and therapeutic delivery strategies. Here, we demonstrate a versatile approach for spatially-defined presentation of engineered proteins within hydrogels through i) immobilization using bio-orthogonal strain-promoted alkyne-azide click chemistry and ii) dynamic protease-driven protein release using exogenously applied enzyme. Model fluorescent proteins were expressed using nonsense codon replacement to incorporate azide-containing unnatural amino acids in a site-specific manner toward maintaining protein activity: here, cyan fluorescent protein (AzCFP), mCherry fluorescent protein (AzmCh), and mCh decorated with a thrombin cut-site. (AzTMBmCh). Eight-arm poly(ethylene glycol) (PEG) was modified with dibenzylcyclooctyne (DBCO) groups and reacted with azide functionalized PEG in aqueous solution for rapid formation of hydrogels. Azide functionalized full-length fluorescent proteins were successfully incorporated into the hydrogel network by reaction with PEG-DBCO prior to gel formation. Temporal release and removal of select proteins (AzTMBmCh) was triggered with the application of thrombin and monitored in real-time with confocal microscopy, providing a responsive handle for controlling matrix properties. Hydrogels with regions of different protein compositions were created using a layering technique with thicknesses of hundreds of micrometers, affording opportunities for the creation of complex geometries on size scales relevant for controlling cellular microenvironments. STATEMENT OF SIGNIFICANCE Controlling protein presentation within biomaterials is important for modulating interactions with biological systems. For example, native tissues are composed of subunits with different matrix compositions (proteins, stiffness) that dynamically interact with cells, influencing function and fate. Toward mimicking such temporally-regulated and spatially-defined microenvironments, we utilize bio-orthogonal click chemistry and protein engineering to create hydrogels with distinct regions of proteins and modify them over time. Through nonsense codon replacement, we site-specifically functionalize large proteins with i) azides for covalent conjugation and ii) an enzymatic cleavage site for user-defined release from hydrogels. Our results exemplify not only the ability to create unique bio-functionalized hydrogels with controlled mechanical properties, but also the potential for creating interesting interfaces for cell culture and tissue engineering applications.
Collapse
Affiliation(s)
- Chen Guo
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Heejae Kim
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Elisa M Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Christine M Mourafetis
- Department of Chemical and Biomolecular Engineering, New York University, Brooklyn, NY 11201, United States
| | - Mingrui Yang
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
144
|
Liu Y, Gill E, Shery Huang YY. Microfluidic on-chip biomimicry for 3D cell culture: a fit-for-purpose investigation from the end user standpoint. Future Sci OA 2017; 3:FSO173. [PMID: 28670465 PMCID: PMC5481809 DOI: 10.4155/fsoa-2016-0084] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/19/2017] [Indexed: 12/13/2022] Open
Abstract
A plethora of 3D and microfluidics-based culture models have been demonstrated in the recent years with the ultimate aim to facilitate predictive in vitro models for pharmaceutical development. This article summarizes to date the progress in the microfluidics-based tissue culture models, including organ-on-a-chip and vasculature-on-a-chip. Specific focus is placed on addressing the question of what kinds of 3D culture and system complexities are deemed desirable by the biological and biomedical community. This question is addressed through analysis of a research survey to evaluate the potential use of microfluidic cell culture models among the end users. Our results showed a willingness to adopt 3D culture technology among biomedical researchers, although a significant gap still exists between the desired systems and existing 3D culture options. With these results, key challenges and future directions are highlighted.
Collapse
Affiliation(s)
- Ye Liu
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, UK, CB2 1PZ
| | - Elisabeth Gill
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, UK, CB2 1PZ
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, UK, CB2 1PZ
| |
Collapse
|
145
|
Fisher SA, Baker AEG, Shoichet MS. Designing Peptide and Protein Modified Hydrogels: Selecting the Optimal Conjugation Strategy. J Am Chem Soc 2017; 139:7416-7427. [PMID: 28481537 DOI: 10.1021/jacs.7b00513] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hydrogels are used in a wide variety of biomedical applications including tissue engineering, biomolecule delivery, cell delivery, and cell culture. These hydrogels are often designed with a specific biological function in mind, requiring the chemical incorporation of bioactive factors to either mimic extracellular matrix or to deliver a payload to diseased tissue. Appropriate synthetic techniques to ligate bioactive factors, such as peptides and proteins, onto hydrogels are critical in designing materials with biological function. Here, we outline strategies for peptide and protein immobilization. We specifically focus on click chemistry, enzymatic ligation, and affinity binding for transient immobilization. Protein modification strategies have shifted toward site-specific modification using unnatural amino acids and engineered site-selective amino acid sequences to preserve both activity and structure. The selection of appropriate protein immobilization strategies is vital to engineering functional hydrogels. We provide insight into chemistry that balances the need for facile reactions while maintaining protein bioactivity or desired release.
Collapse
Affiliation(s)
- Stephanie A Fisher
- The Donnelly Centre for Cellular and Biomolecular Research, ‡Department of Chemical Engineering and Applied Chemistry, §Institute of Biomaterials and Biomedical Engineering, and ∥Department of Chemistry, University of Toronto , 160 College Street, Room 514, Toronto, Ontario M5S 3E1, Canada
| | - Alexander E G Baker
- The Donnelly Centre for Cellular and Biomolecular Research, ‡Department of Chemical Engineering and Applied Chemistry, §Institute of Biomaterials and Biomedical Engineering, and ∥Department of Chemistry, University of Toronto , 160 College Street, Room 514, Toronto, Ontario M5S 3E1, Canada
| | - Molly S Shoichet
- The Donnelly Centre for Cellular and Biomolecular Research, ‡Department of Chemical Engineering and Applied Chemistry, §Institute of Biomaterials and Biomedical Engineering, and ∥Department of Chemistry, University of Toronto , 160 College Street, Room 514, Toronto, Ontario M5S 3E1, Canada
| |
Collapse
|
146
|
Abstract
Hydrogels are formed from hydrophilic polymer chains surrounded by a water-rich environment. They have widespread applications in various fields such as biomedicine, soft electronics, sensors, and actuators. Conventional hydrogels usually possess limited mechanical strength and are prone to permanent breakage. Further, the lack of dynamic cues and structural complexity within the hydrogels has limited their functions. Recent developments include engineering hydrogels that possess improved physicochemical properties, ranging from designs of innovative chemistries and compositions to integration of dynamic modulation and sophisticated architectures. We review major advances in designing and engineering hydrogels and strategies targeting precise manipulation of their properties across multiple scales.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
147
|
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: 108] [Impact Index Per Article: 15.4] [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
|
148
|
Tam RY, Smith LJ, Shoichet MS. Engineering Cellular Microenvironments with Photo- and Enzymatically Responsive Hydrogels: Toward Biomimetic 3D Cell Culture Models. Acc Chem Res 2017; 50:703-713. [PMID: 28345876 DOI: 10.1021/acs.accounts.6b00543] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Conventional cell culture techniques using 2D polystyrene or glass have provided great insight into key biochemical mechanisms responsible for cellular events such as cell proliferation, differentiation, and cell-cell interactions. However, the physical and chemical properties of 2D culture in vitro are dramatically different than those found in the native cellular microenvironment in vivo. Cells grown on 2D substrates differ significantly from those grown in vivo, and this explains, in part, why many promising drug candidates discovered through in vitro drug screening assays fail when they are translated to in vivo animal or human models. To overcome this obstacle, 3D cell culture using biomimetic hydrogels has emerged as an alternative strategy to recapitulate native cell growth in vitro. Hydrogels, which are water-swollen polymers, can be synthetic or naturally derived. Many methods have been developed to control the physical and chemical properties of the hydrogels to match those found in specific tissues. Compared to 2D culture, cells cultured in 3D gels with the appropriate physicochemical cues can behave more like they naturally do in vivo. While conventional hydrogels involve modifications to the bulk material to mimic the static aspects of the cellular microenvironment, recent progress has focused on using more dynamic hydrogels, the chemical and physical properties of which can be altered with external stimuli to better mimic the dynamics of the native cellular microenvironment found in vivo. In this Account, we describe our progress in designing stimuli-responsive, optically transparent hydrogels that can be used as biomimetic extracellular matrices (ECMs) to study cell differentiation and migration in the context of modeling the nervous system and cancer. Specifically, we developed photosensitive agarose and hyaluronic acid hydrogels that are activated by single or two-photon irradiation for biomolecule immobilization at specific volumes within the 3D hydrogel. By controlling the spatial location of protein immobilization, we created 3D patterns and protein concentration gradients within these gels. We used the latter to study the effect of VEGF-165 concentration gradients on the interactions between endothelial cells and retinal stem cells. Hyaluronic acid (HA) is particularly compelling as it is naturally found in the ECM of many tissues and the tumor microenvironment. We used Diels-Alder click chemistry and cryogelation to alter the chemical and physical properties of these hydrogels. We also designed HA hydrogels to study the invasion of breast cancer cells. HA gels were chemically cross-linked with matrix metalloproteinase (MMP)-degradable peptides that degrade in the presence of cancer cell-secreted MMPs, thus allowing cells to remodel their local microenvironment and invade into HA/MMP-degradable gels.
Collapse
Affiliation(s)
- Roger Y. Tam
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Laura J. Smith
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Molly S. Shoichet
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
149
|
Mao M, He J, Li X, Zhang B, Lei Q, Liu Y, Li D. The Emerging Frontiers and Applications of High-Resolution 3D Printing. MICROMACHINES 2017. [PMCID: PMC6190223 DOI: 10.3390/mi8040113] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Mao Mao
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China; (M.M.); (B.Z.); (Q.L.); (Y.L.); (D.L.)
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China; (M.M.); (B.Z.); (Q.L.); (Y.L.); (D.L.)
- Correspondence: ; Tel.: +86-29-83395387
| | - Xiao Li
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA;
| | - Bing Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China; (M.M.); (B.Z.); (Q.L.); (Y.L.); (D.L.)
| | - Qi Lei
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China; (M.M.); (B.Z.); (Q.L.); (Y.L.); (D.L.)
| | - Yaxiong Liu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China; (M.M.); (B.Z.); (Q.L.); (Y.L.); (D.L.)
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China; (M.M.); (B.Z.); (Q.L.); (Y.L.); (D.L.)
| |
Collapse
|
150
|
Zhang N, Milleret V, Thompson-Steckel G, Huang NP, Vörös J, Simona BR, Ehrbar M. Soft Hydrogels Featuring In-Depth Surface Density Gradients for the Simple Establishment of 3D Tissue Models for Screening Applications. SLAS DISCOVERY 2017; 22:635-644. [PMID: 28277889 DOI: 10.1177/2472555217693191] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Three-dimensional (3D) cell culture models are gaining increasing interest for use in drug development pipelines due to their closer resemblance to human tissues. Hydrogels are the first-choice class of materials to recreate in vitro the 3D extra-cellular matrix (ECM) environment, important in studying cell-ECM interactions and 3D cellular organization and leading to physiologically relevant in vitro tissue models. Here we propose a novel hydrogel platform consisting of a 96-well plate containing pre-cast synthetic PEG-based hydrogels for the simple establishment of 3D (co-)culture systems without the need for the standard encapsulation method. The in-depth density gradient at the surface of the hydrogel promotes the infiltration of cells deposited on top of it. The ability to decouple hydrogel production and cell seeding is intended to simplify the use of hydrogel-based platforms and thus increase their accessibility. Using this platform, we established 3D cultures relevant for studying stem cell differentiation, angiogenesis, and neural and cancer models.
Collapse
Affiliation(s)
- Ning Zhang
- 1 Laboratory of Biosensors and Bioelectronics, University and ETH Zurich, Zurich, Switzerland.,2 State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Vincent Milleret
- 3 Laboratory for Cell and Tissue Engineering, Department of Obstetrics, University Hospital Zurich, Zurich, Switzerland.,4 Ectica Technologies AG, Zurich, Switzerland
| | - Greta Thompson-Steckel
- 1 Laboratory of Biosensors and Bioelectronics, University and ETH Zurich, Zurich, Switzerland
| | - Ning-Ping Huang
- 2 State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - János Vörös
- 1 Laboratory of Biosensors and Bioelectronics, University and ETH Zurich, Zurich, Switzerland
| | | | - Martin Ehrbar
- 3 Laboratory for Cell and Tissue Engineering, Department of Obstetrics, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|