1
|
Castillo-Díaz LA, Gough JE, Miller AF, Saiani A. RGD-functionalised self-assembling peptide hydrogel induces a proliferative profile in human osteoblasts in vitro. J Pept Sci 2025; 31:e3653. [PMID: 39329311 DOI: 10.1002/psc.3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024]
Abstract
Self-assembling peptide hydrogels (SAPHs) have been used in the past decade as reliable three-dimensional (3D) synthetic scaffolds for the culture of a variety of mammalian cells in vitro. Thanks to their versatile physicochemical properties, they allow researchers to tailor the hydrogel properties, including stiffness and functionality to the targeted cells and cells' behaviour. One of the advantages of using SAPH scaffolds is the ease of functionalisation. In the present work, we discuss the effect that functionalising the FEFEFKFK (F, phenylalanine; K, lysine; and E, glutamic acid) hydrogel scaffold using the cell-binding RGDS (fibronectin - R, arginine; G, glycine; D, aspartic acid; S, serine) epitope affects the material properties as well as the function of encapsulated human osteoblast cells. RGDS functionalisation resulted in cells adopting an elongated morphology, suggesting attachment and increased proliferation. While this led to higher cell viability, it also resulted in a decrease in extra-cellular matrix (ECM) protein production as well as a decrease in calcium ion deposition, suggesting lower mineralisation capabilities. The work clearly shows that SAPHs are a flexible platform that allow the modification of scaffolds in a controlled manner to investigate cell-material interactions.
Collapse
Affiliation(s)
- Luis A Castillo-Díaz
- School of Chemical Engineering & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
- Departamento de Medicina y Ciencias de la Salud, Facultad Interdisciplinaria de Ciencias Biológicas y de la Salud, Universidad de Sonora, Hermosillo, Sonora, Mexico
| | - Julie E Gough
- School of Materials & Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Aline F Miller
- School of Chemical Engineering & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Alberto Saiani
- Division of Pharmacy and Optometry & Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Liu Y, Gilchrist AE, Heilshorn SC. Engineered Protein Hydrogels as Biomimetic Cellular Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407794. [PMID: 39233559 PMCID: PMC11573243 DOI: 10.1002/adma.202407794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/01/2024] [Indexed: 09/06/2024]
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) play a pivotal role in regulating cellular behaviors such as proliferation, migration, and differentiation. Engineered protein-based hydrogels, with highly tunable multifunctional properties, have the potential to replicate key features of the native ECM. Formed by self-assembly or crosslinking, engineered protein-based hydrogels can induce a range of cell behaviors through bioactive and functional domains incorporated into the polymer backbone. Using recombinant techniques, the amino acid sequence of the protein backbone can be designed with precise control over the chain-length, folded structure, and cell-interaction sites. In this review, the modular design of engineered protein-based hydrogels from both a molecular- and network-level perspective are discussed, and summarize recent progress and case studies to highlight the diverse strategies used to construct biomimetic scaffolds. This review focuses on amino acid sequences that form structural blocks, bioactive blocks, and stimuli-responsive blocks designed into the protein backbone for highly precise and tunable control of scaffold properties. Both physical and chemical methods to stabilize dynamic protein networks with defined structure and bioactivity for cell culture applications are discussed. Finally, a discussion of future directions of engineered protein-based hydrogels as biomimetic cellular scaffolds is concluded.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis 451 Health Sciences Dr, GBSF 3315, Davis, CA, 95616, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, 476 Lomita Mall, McCullough Room 246, Stanford, CA, 94305, USA
| |
Collapse
|
3
|
Liu Y, Gao Z, Yu X, Lin W, Lian H, Meng Z. Recent Advances in the Fabrication and Performance Optimization of Polyvinyl Alcohol Based Vascular Grafts. Macromol Biosci 2024; 24:e2400093. [PMID: 38801024 DOI: 10.1002/mabi.202400093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/11/2024] [Indexed: 05/29/2024]
Abstract
Cardiovascular disease is one of the diseases with the highest morbidity and mortality rates worldwide, and coronary artery bypass grafting (CABG) is a fast and effective treatment. More researchers are investigating in artificial blood vessels due to the limitations of autologous blood vessels. Despite the availability of large-diameter vascular grafts (Ø > 6 mm) for clinical use, small-diameter vascular grafts (Ø < 6 mm) have been a challenge for researchers to overcome in recent years. Vascular grafts made of polyvinyl alcohol (PVA) and PVA-based composites have excellent biocompatibility and mechanical characteristics. In order to gain a clearer and more specific understanding of the progress in PVA vascular graft research, particularly regarding the preparation methods, principles, and functionality of PVA vascular graft, this article discusses the mechanical properties, biocompatibility, blood compatibility, and other properties of PVA vascular graft prepared or enhanced with different blends using various techniques that mimic natural blood vessels. The findings reveal the feasibility and promising potential of PVA or PVA-based composite materials as vascular grafts.
Collapse
Affiliation(s)
- Yixuan Liu
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zichun Gao
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xinrong Yu
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenjiao Lin
- Qingmao Technology (Shenzhen) Co., LTD, Shenzhen, China
| | - He Lian
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhaoxu Meng
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
4
|
Jafari A. Advancements in self-assembling peptides: Bridging gaps in 3D cell culture and electronic device fabrication. J Biomater Appl 2024; 38:1013-1035. [PMID: 38502905 PMCID: PMC11055414 DOI: 10.1177/08853282241240139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Self-assembling peptides (SAPs) show promise in creating synthetic microenvironments that regulate cellular function and tissue repair. Also, the precise π-π interactions and hydrogen bonding within self-assembled peptide structures enable the creation of quantum confined structures, leading to reduced band gaps and the emergence of semiconductor properties within the superstructures. This review emphasizes the need for standardized 3D cell culture methods and electronic devices based on SAPs for monitoring cell communication and controlling cell surface morphology. Additionally, the gap in understanding the relationship between SAP peptide sequences and nanostructures is highlighted, underscoring the importance of optimizing peptide deposition parameters, which affect charge transport and bioactivity due to varying morphologies. The potential of peptide nanofibers as extracellular matrix mimics and the introduction of the zone casting method for improved film deposition are discussed within this review, aiming to bridge knowledge gaps and offer insights into fields like tissue engineering and materials science, with the potential for groundbreaking applications at the interface of biology and materials engineering.
Collapse
Affiliation(s)
- Azadeh Jafari
- Faculty of Applied Sciences, Simon Fraser University, Surrey, BC, Canada
| |
Collapse
|
5
|
Suhar RA, Huang MS, Navarro RS, Aviles Rodriguez G, Heilshorn SC. A Library of Elastin-like Proteins with Tunable Matrix Ligands for In Vitro 3D Neural Cell Culture. Biomacromolecules 2023; 24:5926-5939. [PMID: 37988588 DOI: 10.1021/acs.biomac.3c00941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Hydrogels with encapsulated cells have widespread biomedical applications, both as tissue-mimetic 3D cultures in vitro and as tissue-engineered therapies in vivo. Within these hydrogels, the presentation of cell-instructive extracellular matrix (ECM)-derived ligands and matrix stiffness are critical factors known to influence numerous cell behaviors. While individual ECM biopolymers can be blended together to alter the presentation of cell-instructive ligands, this typically results in hydrogels with a range of mechanical properties. Synthetic systems that allow for the facile incorporation and modulation of multiple ligands without modification of matrix mechanics are highly desirable. In the present work, we leverage protein engineering to design a family of xeno-free hydrogels (i.e., devoid of animal-derived components) consisting of recombinant hyaluronan and recombinant elastin-like proteins (ELPs), cross-linked together with dynamic covalent bonds. The ELP components incorporate cell-instructive peptide ligands derived from ECM proteins, including fibronectin (RGD), laminin (IKVAV and YIGSR), collagen (DGEA), and tenascin-C (PLAEIDGIELTY and VFDNFVL). By carefully designing the protein primary sequence, we form 3D hydrogels with defined and tunable concentrations of cell-instructive ligands that have similar matrix mechanics. Utilizing this system, we demonstrate that neurite outgrowth from encapsulated embryonic dorsal root ganglion (DRG) cultures is significantly modified by cell-instructive ligand content. Thus, this library of protein-engineered hydrogels is a cell-compatible system to systematically study cell responses to matrix-derived ligands.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- The Institute for Chemistry, Stanford University, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford, California 94305, United States
| | - Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Giselle Aviles Rodriguez
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
6
|
Thede AT, Tang JD, Cocker CE, Harold LJ, Amelung CD, Kittel AR, Taylor PA, Lampe KJ. Effects of Cell-Adhesive Ligand Presentation on Pentapeptide Supramolecular Assembly and Gelation: Simulations and Experiments. Cells Tissues Organs 2023; 212:468-483. [PMID: 37751723 DOI: 10.1159/000534280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The extracellular matrix (ECM) is a complex, hierarchical material containing structural and bioactive components. This complexity makes decoupling the effects of biomechanical properties and cell-matrix interactions difficult, especially when studying cellular processes in a 3D environment. Matrix mechanics and cell adhesion are both known regulators of specific cellular processes such as stem cell proliferation and differentiation. However, more information is required about how such variables impact various neural lineages that could, upon transplantation, therapeutically improve neural function after a central nervous system injury or disease. Rapidly Assembling Pentapeptides for Injectable Delivery (RAPID) hydrogels are one biomaterial approach to meet these goals, consisting of a family of peptide sequences that assemble into physical hydrogels in physiological media. In this study, we studied our previously reported supramolecularly-assembling RAPID hydrogels functionalized with the ECM-derived cell-adhesive peptide ligands RGD, IKVAV, and YIGSR. Using molecular dynamics simulations and experimental rheology, we demonstrated that these integrin-binding ligands at physiological concentrations (3-12 mm) did not impact the assembly of the KYFIL peptide system. In simulations, molecular measures of assembly such as hydrogen bonding and pi-pi interactions appeared unaffected by cell-adhesion sequence or concentration. Visualizations of clustering and analysis of solvent-accessible surface area indicated that the integrin-binding domains remained exposed. KYFIL or AYFIL hydrogels containing 3 mm of integrin-binding domains resulted in mechanical properties consistent with their non-functionalized equivalents. This strategy of doping RAPID gels with cell-adhesion sequences allows for the precise tuning of peptide ligand concentration, independent of the rheological properties. The controllability of the RAPID hydrogel system provides an opportunity to investigate the effect of integrin-binding interactions on encapsulated neural cells to discern how hydrogel microenvironment impacts growth, maturation, or differentiation.
Collapse
Affiliation(s)
- Andrew T Thede
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - James D Tang
- University of Virginia Chemical Engineering, Charlottesville, Virginia, USA
| | - Clare E Cocker
- University of Virginia Chemical Engineering, Charlottesville, Virginia, USA
| | - Liza J Harold
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - Connor D Amelung
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - Anna R Kittel
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - Phillip A Taylor
- University of Virginia Chemical Engineering, Charlottesville, Virginia, USA
| | | |
Collapse
|
7
|
Ligorio C, Mata A. Synthetic extracellular matrices with function-encoding peptides. NATURE REVIEWS BIOENGINEERING 2023; 1:1-19. [PMID: 37359773 PMCID: PMC10127181 DOI: 10.1038/s44222-023-00055-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 06/28/2023]
Abstract
The communication of cells with their surroundings is mostly encoded in the epitopes of structural and signalling proteins present in the extracellular matrix (ECM). These peptide epitopes can be incorporated in biomaterials to serve as function-encoding molecules to modulate cell-cell and cell-ECM interactions. In this Review, we discuss natural and synthetic peptide epitopes as molecular tools to bioengineer bioactive hydrogel materials. We present a library of functional peptide sequences that selectively communicate with cells and the ECM to coordinate biological processes, including epitopes that directly signal to cells, that bind ECM components that subsequently signal to cells, and that regulate ECM turnover. We highlight how these epitopes can be incorporated in different biomaterials as individual or multiple signals, working synergistically or additively. This molecular toolbox can be applied in the design of biomaterials aimed at regulating or controlling cellular and tissue function, repair and regeneration.
Collapse
Affiliation(s)
- Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, UK
- School of Pharmacy, University of Nottingham, Nottingham, UK
| |
Collapse
|
8
|
Clever Experimental Designs: Shortcuts for Better iPSC Differentiation. Cells 2021; 10:cells10123540. [PMID: 34944048 PMCID: PMC8700474 DOI: 10.3390/cells10123540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/18/2022] Open
Abstract
For practical use of pluripotent stem cells (PSCs) for disease modelling, drug screening, and regenerative medicine, the cell differentiation process needs to be properly refined to generate end products with consistent and high quality. To construct and optimize a robust cell-induction process, a myriad of cell culture conditions should be considered. In contrast to inefficient brute-force screening, statistical design of experiments (DOE) approaches, such as factorial design, orthogonal array design, response surface methodology (RSM), definitive screening design (DSD), and mixture design, enable efficient and strategic screening of conditions in smaller experimental runs through multifactorial screening and/or quantitative modeling. Although DOE has become routinely utilized in the bioengineering and pharmaceutical fields, the imminent need of more detailed cell-lineage specification, complex organoid construction, and a stable supply of qualified cell-derived material requires expedition of DOE utilization in stem cell bioprocessing. This review summarizes DOE-based cell culture optimizations of PSCs, mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), and Chinese hamster ovary (CHO) cells, which guide effective research and development of PSC-derived materials for academic and industrial applications.
Collapse
|
9
|
Lee KH, Kim TH. Recent Advances in Multicellular Tumor Spheroid Generation for Drug Screening. BIOSENSORS 2021; 11:445. [PMID: 34821661 PMCID: PMC8615712 DOI: 10.3390/bios11110445] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 05/12/2023]
Abstract
Multicellular tumor spheroids (MCTs) have been employed in biomedical fields owing to their advantage in designing a three-dimensional (3D) solid tumor model. For controlling multicellular cancer spheroids, mimicking the tumor extracellular matrix (ECM) microenvironment is important to understand cell-cell and cell-matrix interactions. In drug cytotoxicity assessments, MCTs provide better mimicry of conventional solid tumors that can precisely represent anticancer drug candidates' effects. To generate incubate multicellular spheroids, researchers have developed several 3D multicellular spheroid culture technologies to establish a research background and a platform using tumor modelingvia advanced materials science, and biosensing techniques for drug-screening. In application, drug screening was performed in both invasive and non-invasive manners, according to their impact on the spheroids. Here, we review the trend of 3D spheroid culture technology and culture platforms, and their combination with various biosensing techniques for drug screening in the biomedical field.
Collapse
Affiliation(s)
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Korea;
| |
Collapse
|
10
|
Spiller S, Clauder F, Bellmann-Sickert K, Beck-Sickinger AG. Improvement of wound healing by the development of ECM-inspired biomaterial coatings and controlled protein release. Biol Chem 2021; 402:1271-1288. [PMID: 34392636 DOI: 10.1515/hsz-2021-0144] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/19/2021] [Indexed: 12/22/2022]
Abstract
Implant design has evolved from biochemically inert substrates, minimizing cell and protein interaction, towards sophisticated bioactive substrates, modulating the host response and supporting the regeneration of the injured tissue. Important aspects to consider are the control of cell adhesion, the discrimination of bacteria and non-local cells from the desired tissue cell type, and the stimulation of implant integration and wound healing. Here, the extracellular matrix acts as a role model providing us with inspiration for sophisticated designs. Within this scope, small bioactive peptides have proven to be miscellaneously deployable for the mediation of surface, cell and matrix interactions. Combinations of adhesion ligands, proteoglycans, and modulatory proteins should guide multiple aspects of the regeneration process and cooperativity between the different extracellular matrix components, which bears the chance to maximize the therapeutic efficiency and simultaneously lower the doses. Hence, efforts to include multiple of these factors in biomaterial design are well worth. In the following, multifunctional implant coatings based on bioactive peptides are reviewed and concepts to implement strong surface anchoring for stable cell adhesion and a dynamic delivery of modulator proteins are discussed.
Collapse
Affiliation(s)
- Sabrina Spiller
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103Leipzig, Germany
| |
Collapse
|
11
|
van Gaal RC, Vrehen AF, van Sprang JF, Fransen PPKH, van Turnhout MC, Dankers PYW. Biomaterial screening of protein coatings and peptide additives: towards a simple synthetic mimic of a complex natural coating for a bio-artificial kidney. Biomater Sci 2021; 9:2209-2220. [PMID: 33506836 DOI: 10.1039/d0bm01930e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bio-artificial kidneys require conveniently synthesized membranes providing signals that regulate renal epithelial cell function. Therefore, we aimed to find synthetic analogues for natural extracellular matrix (ECM) protein coatings traditionally used for epithelial cell culturing. Two biomaterial libraries, based on natural ECM-coatings and on synthetic supramolecular small molecule additives, were developed. The base material consisted of a bisurea (BU) containing polymer, providing supramolecular BU-additives to be incorporated via specific hydrogen bonding interactions. This system allows for a modular approach and therefore easy fractional factorial based screening. A natural coating on the BU-polymer material with basement membrane proteins, laminin and collagen IV, combined with catechols was shown to induce renal epithelial monolayer formation. Modification of the BU-polymer material with synthetic BU-modified ECM peptide additives did not result in monolayer formation. Unexpectedly, simple BU-catechol additives induced monolayer formation and presented similar levels of epithelial markers and apical transporter function as on the laminin, collagen IV and catechol natural coating. Importantly, when this BU-polymer material was processed into fibrous e-spun membranes the natural coating and the BU-catechol additive were shown to perfectly function. This study clearly indicates that complex natural ECM-coatings can be replaced by simple synthetic additives, and displays the potency of material libraries based on design of experiments in combination with modular, supramolecular chemistry.
Collapse
Affiliation(s)
- Ronald C van Gaal
- Laboratory for Cell and Tissue Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
| | | | | | | | | | | |
Collapse
|
12
|
Yasui R, Sekine K, Yamaguchi K, Furukawa Y, Taniguchi H. Robust parameter design of human induced pluripotent stem cell differentiation protocols defines lineage-specific induction of anterior-posterior gut tube endodermal cells. Stem Cells 2021; 39:429-442. [PMID: 33400835 DOI: 10.1002/stem.3326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022]
Abstract
Tissues and cells derived from pluripotent stem cells (PSC) are likely to become widely used in disease modeling, drug screening, and regenerative medicine. For these applications, the in vitro PSC differentiation process must be elaborately investigated and controlled to reliably obtain the desired end products. However, because traditional experimental methods, such as one factor at a time or brute-force approaches, are impractical for detailed screening of complex PSC cultivation conditions, more strategic and effective screening based on statistical design of experiments (DOE) ought to be indispensable. Among various DOE approaches, we regard robust parameter design (RPD) as particularly suited for differentiation protocol optimization due to its suitability for multifactorial screening. We confirmed the adaptability of RPD for investigating human induced PSC lineage specification toward anterior-posterior gut tube endodermal cells and clarified both the contribution of each cell signaling pathway and the effect of cell signaling condition alteration on marker RNA expression levels, while increasing the efficiency of the screening in 243-fold (18 vs 4374) compared with that of a brute-force approach. Specific induction of anterior foregut, hepatic, pancreatic, or mid-hindgut cells was achieved using seven iPSC strains with the optimal culture protocols established on the basis of RPD analysis. RPD has the potential to enable efficient construction and optimization of PSC differentiation protocols, and its use is recommended from fundamental research to mass production of PSC-derived products.
Collapse
Affiliation(s)
- Ryota Yasui
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Fundamental Research Laboratory, Eiken Chemical Co., Ltd., Nogi, Tochigi, Japan
| | - Keisuke Sekine
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Cancer Cell Systems, National Cancer Center Research Institute, Tokyo, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.,Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Advanced Medical Research Center, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
13
|
Abstract
Self-assembly of proteins and peptides into the amyloid fold is a widespread phenomenon in the natural world. The structural hallmark of self-assembly into amyloid fibrillar assemblies is the cross-beta motif, which conveys distinct morphological and mechanical properties. The amyloid fibril formation has contrasting results depending on the organism, in the sense that it can bestow an organism with the advantages of mechanical strength and improved functionality or, on the contrary, could give rise to pathological states. In this chapter we review the existing information on amyloid-like peptide aggregates, which could either be derived from protein sequences, but also could be rationally or de novo designed in order to self-assemble into amyloid fibrils under physiological conditions. Moreover, the development of self-assembled fibrillar biomaterials that are tailored for the desired properties towards applications in biomedical or environmental areas is extensively analyzed. We also review computational studies predicting the amyloid propensity of the natural amino acid sequences and the structure of amyloids, as well as designing novel functional amyloid materials.
Collapse
Affiliation(s)
- C. Kokotidou
- University of Crete, Department of Materials Science and Technology Voutes Campus GR-70013 Heraklion Crete Greece
- FORTH, Institute for Electronic Structure and Laser N. Plastira 100 GR 70013 Heraklion Greece
| | - P. Tamamis
- Texas A&M University, Artie McFerrin Department of Chemical Engineering College Station Texas 77843-3122 USA
| | - A. Mitraki
- University of Crete, Department of Materials Science and Technology Voutes Campus GR-70013 Heraklion Crete Greece
- FORTH, Institute for Electronic Structure and Laser N. Plastira 100 GR 70013 Heraklion Greece
| |
Collapse
|
14
|
Shores LS, Kelly SH, Hainline KM, Suwanpradid J, MacLeod AS, Collier JH. Multifactorial Design of a Supramolecular Peptide Anti-IL-17 Vaccine Toward the Treatment of Psoriasis. Front Immunol 2020; 11:1855. [PMID: 32973764 PMCID: PMC7461889 DOI: 10.3389/fimmu.2020.01855] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
Current treatments for chronic immune-mediated diseases such as psoriasis, rheumatoid arthritis, or Crohn's disease commonly rely on cytokine neutralization using monoclonal antibodies; however, such approaches have drawbacks. Frequent repeated dosing can lead to the formation of anti-drug antibodies and patient compliance issues, and it is difficult to identify a single antibody that is broadly efficacious across diverse patient populations. As an alternative to monoclonal antibody therapy, anti-cytokine immunization is a potential means for long-term therapeutic control of chronic inflammatory diseases. Here we report a supramolecular peptide-based approach for raising antibodies against IL-17 and demonstrate its efficacy in a murine model of psoriasis. B-cell epitopes from IL-17 were co-assembled with the universal T-cell epitope PADRE using the Q11 self-assembling peptide nanofiber system. These materials, with or without adjuvants, raised antibody responses against IL-17. Exploiting the modularity of the system, multifactorial experimental designs were used to select formulations maximizing titer and avidity. In a mouse model of psoriasis induced by imiquimod, unadjuvanted nanofibers had therapeutic efficacy, which could be enhanced with alum adjuvant but reversed with CpG adjuvant. Measurements of antibody subclass induced by adjuvanted and unadjuvanted formulations revealed strong correlations between therapeutic efficacy and titers of IgG1 (improved efficacy) or IgG2b (worsened efficacy). These findings have important implications for the development of anti-cytokine active immunotherapies and suggest that immune phenotype is an important metric for eliciting therapeutic anti-cytokine antibody responses.
Collapse
Affiliation(s)
- Lucas S Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Sean H Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Jutamas Suwanpradid
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University School of Medicine, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
15
|
Dickmeis C, Kauth L, Commandeur U. From infection to healing: The use of plant viruses in bioactive hydrogels. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1662. [PMID: 32677315 DOI: 10.1002/wnan.1662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/08/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022]
Abstract
Plant viruses show great diversity in shape and size, but each species forms unique nucleoprotein particles that are symmetrical and monodisperse. The genetically programed structure of plant viruses allows them to be modified by genetic engineering, bioconjugation, or encapsulation to form virus nanoparticles (VNPs) that are suitable for a broad range of applications. Plant VNPs can be used to present foreign proteins or epitopes, to construct inorganic hybrid materials, or to carry molecular cargos, allowing their utilization as imaging reagents, immunomodulators, therapeutics, nanoreactors, and biosensors. The medical applications of plant viruses benefit from their inability to infect and replicate in human cells. The structural properties of plant viruses also make them useful as components of hydrogels for tissue engineering. Hydrogels are three-dimensional networks composed of hydrophilic polymers that can absorb large amounts of water. They are used as supports for tissue regeneration, as reservoirs for controlled drug release, and are found in contact lenses, many wound healing materials, and hygiene products. They are also useful in ecological applications such as wastewater treatment. Hydrogel-based matrices are structurally similar to the native extracellular matrix (ECM) and provide a scaffold for the attachment of cells. To fully replicate the functions of the ECM it is necessary to augment hydrogels with biological cues that regulate cellular interactions. This can be achieved by incorporating functionalized VNPs displaying ligands that influence the mechanical characteristics of hydrogels and their biological properties, promoting the survival, proliferation, migration, and differentiation of embedded cells. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Christina Dickmeis
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Louisa Kauth
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Ulrich Commandeur
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
16
|
Marquardt LM, Doulames VM, Wang AT, Dubbin K, Suhar RA, Kratochvil MJ, Medress ZA, Plant GW, Heilshorn SC. Designer, injectable gels to prevent transplanted Schwann cell loss during spinal cord injury therapy. SCIENCE ADVANCES 2020; 6:eaaz1039. [PMID: 32270042 PMCID: PMC7112763 DOI: 10.1126/sciadv.aaz1039] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/08/2020] [Indexed: 05/09/2023]
Abstract
Transplantation of patient-derived Schwann cells is a promising regenerative medicine therapy for spinal cord injuries; however, therapeutic efficacy is compromised by inefficient cell delivery. We present a materials-based strategy that addresses three common causes of transplanted cell death: (i) membrane damage during injection, (ii) cell leakage from the injection site, and (iii) apoptosis due to loss of endogenous matrix. Using protein engineering and peptide-based assembly, we designed injectable hydrogels with modular cell-adhesive and mechanical properties. In a cervical contusion model, our hydrogel matrix resulted in a greater than 700% improvement in successful Schwann cell transplantation. The combination therapy of cells and gel significantly improved the spatial distribution of transplanted cells within the endogenous tissue. A reduction in cystic cavitation and neuronal loss were also observed with substantial increases in forelimb strength and coordination. Using an injectable hydrogel matrix, therefore, can markedly improve the outcomes of cellular transplantation therapies.
Collapse
Affiliation(s)
- Laura M. Marquardt
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vanessa M. Doulames
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alice T. Wang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Karen Dubbin
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Riley A. Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Michael J. Kratochvil
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
- Division of Infectious Diseases, Stanford University School of Medicine, Stanford CA 94305, USA
| | - Zachary A. Medress
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Giles W. Plant
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Corresponding author. (G.W.P.); (S.C.H.)
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
- Corresponding author. (G.W.P.); (S.C.H.)
| |
Collapse
|
17
|
He YJ, Santana MF, Moucka M, Quirk J, Shuaibi A, Pimentel MB, Grossman S, Rashid MM, Cinar A, Georgiadis JG, Vaicik M, Kawaji K, Venerus DC, Papavasiliou G. Immobilized RGD concentration and proteolytic degradation synergistically enhance vascular sprouting within hydrogel scaffolds of varying modulus. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2020; 31:324-349. [PMID: 31774730 PMCID: PMC7185153 DOI: 10.1080/09205063.2019.1692640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/30/2019] [Accepted: 11/11/2019] [Indexed: 12/19/2022]
Abstract
Insufficient vascularization limits the volume and complexity of engineered tissue. The formation of new blood vessels (neovascularization) is regulated by a complex interplay of cellular interactions with biochemical and biophysical signals provided by the extracellular matrix (ECM) necessitating the development of biomaterial approaches that enable systematic modulation in matrix properties. To address this need poly(ethylene) glycol-based hydrogel scaffolds were engineered with a range of decoupled and combined variations in integrin-binding peptide (RGD) ligand concentration, elastic modulus and proteolytic degradation rate using free-radical polymerization chemistry. The modularity of this system enabled a full factorial experimental design to simultaneously investigate the individual and interaction effects of these matrix cues on vascular sprout formation in 3 D culture. Enhancements in scaffold proteolytic degradation rate promoted significant increases in vascular sprout length and junction number while increases in modulus significantly and negatively impacted vascular sprouting. We also observed that individual variations in immobilized RGD concentration did not significantly impact 3 D vascular sprouting. Our findings revealed a previously unidentified and optimized combination whereby increases in both immobilized RGD concentration and proteolytic degradation rate resulted in significant and synergistic enhancements in 3 D vascular spouting. The above-mentioned findings would have been challenging to uncover using one-factor-at-time experimental analyses.
Collapse
Affiliation(s)
- Yusheng J. He
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Martin F. Santana
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Madison Moucka
- Department of Biomedical Engineering, Texas A & M University, College Station, TX
| | - Jack Quirk
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Asma Shuaibi
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Marja B. Pimentel
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Sophie Grossman
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Mudassir M. Rashid
- Department Chemical and Biological Engineering Department, Illinois Institute of Technology, Chicago, IL
| | - Ali Cinar
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
- Department Chemical and Biological Engineering Department, Illinois Institute of Technology, Chicago, IL
| | - John G. Georgiadis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Marcella Vaicik
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - Keigo Kawaji
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| | - David C. Venerus
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ
| | - Georgia Papavasiliou
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL
| |
Collapse
|
18
|
Ippel BD, Arts B, Keizer HM, Dankers PYW. Combinatorial functionalization with bisurea-peptides and antifouling bisurea additives of a supramolecular elastomeric biomaterial. JOURNAL OF POLYMER SCIENCE. PART B, POLYMER PHYSICS 2019; 57:1725-1735. [PMID: 32025088 PMCID: PMC6988465 DOI: 10.1002/polb.24907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
The bioactive additive toolbox to functionalize supramolecular elastomeric materials expands rapidly. Here we have set an explorative step toward screening of complex combinatorial functionalization with antifouling and three peptide-containing additives in a bisurea-based supramolecular system. Thorough investigation of surface properties of thin films with contact angle measurements, X-ray photoelectron spectroscopy and atomic force microscopy, was correlated to cell-adhesion of endothelial and smooth muscle cells to apprehend their respective predictive values for functional biomaterial development. Peptides were presented at the surface alone, and in combinatorial functionalization with the oligo(ethylene glycol)-based non-cell adhesive additive. The bisurea-RGD additive was cell-adhesive in all conditions, whereas the endothelial cell-specific bisurea-REDV showed limited bioactive properties in all chemical nano-environments. Also, aspecific functionality was observed for a bisurea-SDF1α peptide. These results emphasize that special care should be taken in changing the chemical nano-environment with peptide functionalization. © 2019 The Authors. Journal of Polymer Science Part B: Polymer Physics published by Wiley Periodicals, Inc. J. Polym. Sci., Part B: Polym. Phys. 2019, 57, 1725-1735.
Collapse
Affiliation(s)
- Bastiaan D. Ippel
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory for Cell and Tissue EngineeringEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
| | - Boris Arts
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical BiologyEindhoven University of TechnologyPO Box 513, 5600EindhovenManitobaThe Netherlands
| | - Henk M. Keizer
- SyMO‐Chem B.VDen Dolech 2, 5612EindhovenArizonaThe Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular SystemsEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory for Cell and Tissue EngineeringEindhoven University of TechnologyPO Box 513 5600EindhovenManitobaThe Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical BiologyEindhoven University of TechnologyPO Box 513, 5600EindhovenManitobaThe Netherlands
| |
Collapse
|
19
|
Clauder F, Czerniak AS, Friebe S, Mayr SG, Scheinert D, Beck-Sickinger AG. Endothelialization of Titanium Surfaces by Bioinspired Cell Adhesion Peptide Coatings. Bioconjug Chem 2019; 30:2664-2674. [DOI: 10.1021/acs.bioconjchem.9b00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Anne Sophie Czerniak
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sabrina Friebe
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Stefan G. Mayr
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Dierk Scheinert
- Department of Angiology, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| |
Collapse
|
20
|
Su J, Satchell SC, Wertheim JA, Shah RN. Poly(ethylene glycol)-crosslinked gelatin hydrogel substrates with conjugated bioactive peptides influence endothelial cell behavior. Biomaterials 2019; 201:99-112. [PMID: 30807988 PMCID: PMC6777960 DOI: 10.1016/j.biomaterials.2019.02.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/28/2022]
Abstract
The basement membrane is a specialized extracellular matrix substrate responsible for support and maintenance of epithelial and endothelial structures. Engineered basement membrane-like hydrogel systems have the potential to advance understanding of cell-cell and cell-matrix interactions by allowing precise tuning of the substrate or matrix biochemical and biophysical properties. In this investigation, we developed tunable hydrogel substrates with conjugated bioactive peptides to modulate cell binding and growth factor signaling by endothelial cells. Hydrogels were formed by employing a poly(ethylene glycol) crosslinker to covalently crosslink gelatin polymers and simultaneously conjugate laminin-derived YIGSR peptides or vascular endothelial growth factor (VEGF)-mimetic QK peptides to the gelatin. Rheological characterization revealed rapid formation of hydrogels with similar stiffnesses across tested formulations, and swelling analysis demonstrated dependency on peptide and crosslinker concentrations in hydrogels. Levels of phosphorylated VEGF Receptor 2 in cells cultured on hydrogel substrates revealed that while human umbilical vein endothelial cells (HUVECs) responded to both soluble and conjugated forms of the QK peptide, conditionally-immortalized human glomerular endothelial cells (GEnCs) only responded to the conjugated presentation of the peptide. Furthermore, whereas HUVECs exhibited greatest upregulation in gene expression when cultured on YIGSR- and QK-conjugated hydrogel substrates after 5 days, GEnCs exhibited greatest upregulation when cultured on Matrigel control substrates at the same time point. These results indicate that conjugation of bioactive peptides to these hydrogel substrates significantly influenced endothelial cell behavior in cultures but with differential responses between HUVECs and GEnCs.
Collapse
Affiliation(s)
- Jimmy Su
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL, USA; Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Simon C Satchell
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Bristol, United Kingdom
| | - Jason A Wertheim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL, USA; Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA; Department of Surgery, Jesse Brown VA Medical Center, Chicago, IL, USA.
| | - Ramille N Shah
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
21
|
Leach DG, Young S, Hartgerink JD. Advances in immunotherapy delivery from implantable and injectable biomaterials. Acta Biomater 2019; 88:15-31. [PMID: 30771535 PMCID: PMC6632081 DOI: 10.1016/j.actbio.2019.02.016] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/10/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
Abstract
Macroscale biomaterials, such as preformed implantable scaffolds and injectable soft materials, possess powerful synergies with anti-cancer immunotherapies. Immunotherapies on their own typically have poor delivery properties, and often require repeated high-dose injections that result in serious off-tumor effects and/or limited efficacy. Rationally designed biomaterials allow for discrete localization and controlled release of immunotherapeutic agents, and have been shown in a large number of applications to improve outcomes in the treatment of cancers via immunotherapy. Among various strategies, macroscale biomaterial delivery systems can take the form of robust tablet-like scaffolds that are surgically implanted into a tumor resection site, releasing programmed immune cells or immunoregulatory agents. Alternatively they can be developed as soft gel-like materials that are injected into solid tumors or sites of resection to stimulate a potent anti-tumor immune response. Biomaterials synthesized from diverse components such as polymers and peptides can be combined with any immunotherapy in the modern toolbox, from checkpoint inhibitors and stimulatory adjuvants, to cancer antigens and adoptive T cells, resulting in unique synergies and improved therapeutic efficacy. The field is growing rapidly in size as publications continue to appear in the literature, and biomaterial-based immunotherapies are entering clinical trials and human patients. It is unarguably an exciting time for cancer immunotherapy and biomaterial researchers, and further work seeks to understand the most critical design considerations in the development of the next-generation of immunotherapeutic biomaterials. This review will discuss recent advances in the delivery of immunotherapies from localized biomaterials, focusing on macroscale implantable and injectable systems. STATEMENT OF SIGNIFICANCE: Anti-cancer immunotherapies have shown exciting clinical results in the past few decades, yet they suffer from a few distinct limitations, such as poor delivery kinetics, narrow patient response profiles, and systemic side effects. Biomaterial systems are now being developed that can overcome many of these problems, allowing for localized adjuvant delivery, focused dose concentrations, and extended therapy presentation. The field of biocompatible carrier materials is uniquely suited to be combined with immunotherapy, promising to yield significant improvements in treatment outcomes and clinical care. In this review, the first pioneering efforts and most recent advances in biomaterials for immunotherapeutic applications are explored, with a specific focus on implantable and injectable biomaterials such as porous scaffolds, cryogels, and hydrogels.
Collapse
Affiliation(s)
- David G Leach
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Simon Young
- Department of Oral & Maxillofacial Surgery, University of Texas Health Science Center, Houston, TX 77054, United States
| | - Jeffrey D Hartgerink
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, TX 77005, United States.
| |
Collapse
|
22
|
Anderson DEJ, Truong KP, Hagen MW, Yim EKF, Hinds MT. Biomimetic modification of poly(vinyl alcohol): Encouraging endothelialization and preventing thrombosis with antiplatelet monotherapy. Acta Biomater 2019; 86:291-299. [PMID: 30639349 DOI: 10.1016/j.actbio.2019.01.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/17/2018] [Accepted: 01/09/2019] [Indexed: 01/04/2023]
Abstract
Poly(vinyl alcohol) (PVA) has shown promise as a biomaterial for cardiovascular application. However, its antifouling properties prevent in vivo endothelialization. This work examined the endothelialization and thrombogenicity of modified PVA with different concentrations of proteins and adhesion peptides: collagen, laminin, fibronectin, GFPGER, YIGSR, and cRGD. Material surface properties were quantified, and the endothelialization potential was determined with human endothelial colony forming cells. Additionally, platelet attachment was assessed in vitro with human platelet rich plasma, and promising samples were tested in an ex vivo shunt model. This well-established arteriovenous shunt model was used with and without clinically-relevant antiplatelet therapies, specifically acetylsalicylic acid (ASA) with and without clopidogrel to examine the minimum necessary treatment to prevent thrombosis. Collagen, laminin, and GFPGER biomolecules increased endothelialization, with GFPGER showing the greatest effect at the lowest concentrations. GFPGER-PVA tubes tested under whole blood did exhibit an increase in platelet (but not fibrin) attachment compared to plain PVA and clinical controls. However, application of ASA monotherapy reduced the thrombogenicity of GFPGER-PVA below the clinical control with the ASA. This work is significant in developing cardiovascular biomaterials-increasing endothelialization potential while reducing bleeding side effects by using an antiplatelet monotherapy, typical of clinical patients. STATEMENT OF SIGNIFICANCE: We modified the endothelialization potential of synthetic, hydrogel vascular grafts with proteins and peptides of the vascular tissue matrix. Cell attachment was dramatically increased with the GFPGER peptide, and while some additional platelet attachment was seen under flow with whole blood, this was completely knocked down using clinical antiplatelet monotherapy. This indicates that long-term patency of this biomaterial could be improved without the associated bleeding risk of multiple platelet therapies.
Collapse
Affiliation(s)
- Deirdre E J Anderson
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave., Portland, OR 97239, USA
| | - Katie P Truong
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave., Portland, OR 97239, USA
| | - Matthew W Hagen
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave., Portland, OR 97239, USA
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, 200 University Ave. W, Waterloo, ON N2L3G1, Canada
| | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 SW Bond Ave., Portland, OR 97239, USA.
| |
Collapse
|
23
|
Hainline KM, Gu F, Handley JF, Tian YF, Wu Y, de Wet L, Vander Griend DJ, Collier JH. Self-Assembling Peptide Gels for 3D Prostate Cancer Spheroid Culture. Macromol Biosci 2018; 19:e1800249. [PMID: 30324687 DOI: 10.1002/mabi.201800249] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Progress in prostate cancer research is presently limited by a shortage of reliable in vitro model systems. The authors describe a novel self-assembling peptide, bQ13, which forms nanofibers and gels useful for the 3D culture of prostate cancer spheroids, with improved cytocompatibility compared to related fibrillizing peptides. The mechanical properties of bQ13 gels can be controlled by adjusting peptide concentration, with storage moduli ranging between 1 and 10 kPa. bQ13's ability to remain soluble at mildly basic pH considerably improved the viability of encapsulated cells compared to other self-assembling nanofiber-forming peptides. LNCaP cells formed spheroids in bQ13 gels with similar morphologies and sizes to those formed in Matrigel or RADA16-I. Moreover, prostate-specific antigen (PSA) is produced by LNCaP cells in all matrices, and PSA production is more responsive to enzalutamide treatment in bQ13 gels than in other fibrillized peptide gels. bQ13 represents an attractive platform for further tailoring within 3D cell culture systems.
Collapse
Affiliation(s)
- Kelly M Hainline
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Fangqi Gu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Jacqueline F Handley
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Ye F Tian
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Larischa de Wet
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Donald J Vander Griend
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL, 60637, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
24
|
Jung JP, Lin WH, Riddle MJ, Tolar J, Ogle BM. A 3D in vitro model of the dermoepidermal junction amenable to mechanical testing. J Biomed Mater Res A 2018; 106:3231-3238. [PMID: 30208260 PMCID: PMC6283247 DOI: 10.1002/jbm.a.36519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/20/2018] [Accepted: 07/31/2018] [Indexed: 01/06/2023]
Abstract
Recessive dystrophic Epidermolysis Bullosa (RDEB) is caused by mutations in collagen‐type VII gene critical for the dermoepidermal junction (DEJ) formation. Neither tissues of animal models nor currently available in vitro models are amenable to the quantitative assessment of mechanical adhesion between dermal and epidermal layers. Here, we created a 3D in vitro DEJ model using extracellular matrix (ECM) proteins of the DEJ anchored to a poly(ethylene glycol)‐based slab (termed ECM composites) and seeded with human keratinocytes and dermal fibroblasts. Keratinocytes and fibroblasts of healthy individuals were well maintained in the ECM composite and showed the expression of collagen type VII over a 2‐week period. The ECM composites with healthy keratinocytes and fibroblasts exhibited yield stress associated with the separation of the model DEJ at 0.268 ± 0.057 kPa. When we benchmarked this measure of adhesive strength with that of the model DEJ fabricated with cells of individuals with RDEB, the yield stress was significantly lower (0.153 ± 0.064 kPa) consistent with our current mechanistic understanding of RDEB. In summary, a 3D in vitro model DEJ was developed for quantification of mechanical adhesion between epidermal‐ and dermal‐mimicking layers, which can be utilized for assessment of mechanical adhesion of the model DEJ applicable for Epidermolysis Bullosa‐associated therapeutics. © 2018 The Authors. Journal Of Biomedical Materials Research Part A Published By Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3231–3238, 2018.
Collapse
Affiliation(s)
- Jangwook P Jung
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana
| | - Wei-Han Lin
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Megan J Riddle
- Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Jakub Tolar
- Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Lillehei Heart Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
25
|
Chesson CB, Huante M, Nusbaum RJ, Walker AG, Clover TM, Chinnaswamy J, Endsley JJ, Rudra JS. Nanoscale Peptide Self-assemblies Boost BCG-primed Cellular Immunity Against Mycobacterium tuberculosis. Sci Rep 2018; 8:12519. [PMID: 30131591 PMCID: PMC6104033 DOI: 10.1038/s41598-018-31089-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/23/2018] [Indexed: 11/30/2022] Open
Abstract
Bacillus Calmette-Guerin (BCG) is the only vaccine against TB and has limited protection efficacy, which wanes past adolescence. Multifunctional CD8+ T cells (IFN-γ+/TNF-α+/IL-2+) are associated with lower reactivation risk and enhanced control of active Mtb infection. Since boosting with BCG is contraindicated, booster vaccines that augment T cell immunity in the lungs of BCG-vaccinated individuals are urgently needed. We developed a vaccination strategy based on self-assembling peptide nanofibers presenting Mtb-specific CD8+ or CD4+ T cell epitopes that induce high frequency and antigen-specific effector memory T cells producing IFN-γ and IL-2. Intranasal immunization with peptide nanofibers was well tolerated in mice leading to increased antigen-specific CD8+ T cell population in the lungs. Co-assembled nanofibers of CD8+ T cell epitopes and toll-like receptor 2 (TLR2) agonists induced a 8-fold expansion in multifunctional CD8+ T cell populations in the lungs of vaccinated mice. Aerosol challenge with Mtb in BCG-primed and nanofiber-boosted mice provided an additional 0.5-log CFU reduction in lung bacterial load and indicating enhanced protection compared to BCG alone. Together, these data suggest that heterologous prime-boost with BCG and peptide nanofiber vaccines induces cell mediated immunity in the lung, reduces bacterial burden, and is a potentially safer alternative for boosting BCG-primed immunity.
Collapse
Affiliation(s)
- Charles B Chesson
- Department of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08823, USA
| | - Matthew Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Rebecca J Nusbaum
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Aida G Walker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, 77555, Texas, USA
| | - Tara M Clover
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, 77555, Texas, USA
| | - Jagannath Chinnaswamy
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Jai S Rudra
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, 77555, Texas, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
26
|
Nguyen EH, Murphy WL. Customizable biomaterials as tools for advanced anti-angiogenic drug discovery. Biomaterials 2018; 181:53-66. [PMID: 30077137 DOI: 10.1016/j.biomaterials.2018.07.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The inhibition of angiogenesis is a critical element of cancer therapy, as cancer vasculature contributes to tumor expansion. While numerous drugs have proven to be effective at disrupting cancer vasculature, patient survival has not significantly improved as a result of anti-angiogenic drug treatment. Emerging evidence suggests that this is due to a combination of unintended side effects resulting from the application of anti-angiogenic compounds, including angiogenic rebound after treatment and the activation of metastasis in the tumor. There is currently a need to better understand the far-reaching effects of anti-angiogenic drug treatments in the context of cancer. Numerous innovations and discoveries in biomaterials design and tissue engineering techniques are providing investigators with tools to develop physiologically relevant vascular models and gain insights into the holistic impact of drug treatments on tumors. This review examines recent advances in the design of pro-angiogenic biomaterials, specifically in controlling integrin-mediated cell adhesion, growth factor signaling, mechanical properties and oxygen tension, as well as the implementation of pro-angiogenic materials into sophisticated co-culture models of cancer vasculature.
Collapse
Affiliation(s)
- Eric H Nguyen
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
27
|
Abstract
Self-assembled peptide nanostructures have been increasingly exploited as functional materials for applications in biomedicine and energy. The emergent properties of these nanomaterials determine the applications for which they can be exploited. It has recently been appreciated that nanomaterials composed of multicomponent coassembled peptides often display unique emergent properties that have the potential to dramatically expand the functional utility of peptide-based materials. This review presents recent efforts in the development of multicomponent peptide assemblies. The discussion includes multicomponent assemblies derived from short low molecular weight peptides, peptide amphiphiles, coiled coil peptides, collagen, and β-sheet peptides. The design, structure, emergent properties, and applications for these multicomponent assemblies are presented in order to illustrate the potential of these formulations as sophisticated next-generation bio-inspired materials.
Collapse
Affiliation(s)
- Danielle M Raymond
- Department of Chemistry, University of Rochester, Rochester, NY 14627-0216, USA.
| | | |
Collapse
|
28
|
Johns M, Bae Y, Guimarães FEG, Lanzoni EM, Costa CAR, Murray PM, Deneke C, Galembeck F, Scott JL, Sharma RI. Predicting Ligand-Free Cell Attachment on Next-Generation Cellulose-Chitosan Hydrogels. ACS OMEGA 2018; 3:937-945. [PMID: 30023793 PMCID: PMC6045362 DOI: 10.1021/acsomega.7b01583] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/02/2018] [Indexed: 06/08/2023]
Abstract
There is a growing appreciation that engineered biointerfaces can regulate cell behaviors, or functions. Most systems aim to mimic the cell-friendly extracellular matrix environment and incorporate protein ligands; however, the understanding of how a ligand-free system can achieve this is limited. Cell scaffold materials comprised of interfused chitosan-cellulose hydrogels promote cell attachment in ligand-free systems, and we demonstrate the role of cellulose molecular weight, MW, and chitosan content and MW in controlling material properties and thus regulating cell attachment. Semi-interpenetrating network (SIPN) gels, generated from cellulose/ionic liquid/cosolvent solutions, using chitosan solutions as phase inversion solvents, were stable and obviated the need for chemical coupling. Interface properties, including surface zeta-potential, dielectric constant, surface roughness, and shear modulus, were modified by varying the chitosan degree of polymerization and solution concentration, as well as the source of cellulose, creating a family of cellulose-chitosan SIPN materials. These features, in turn, affect cell attachment onto the hydrogels and the utility of this ligand-free approach is extended by forecasting cell attachment using regression modeling to isolate the effects of individual parameters in an initially complex system. We demonstrate that increasing the charge density, and/or shear modulus, of the hydrogel results in increased cell attachment.
Collapse
Affiliation(s)
- Marcus
A. Johns
- Department
of Chemical Engineering, Centre for Sustainable Chemical
Technologies, and Department of Chemistry, University of
Bath, Bath BA2 7AY, U.K.
| | - Yongho Bae
- Department
of Pathology and Anatomical Sciences, Jacobs School of Medicine and
Biomedical Sciences, University at Buffalo,
The State University of New York, Buffalo, New York 14203, United States
| | | | - Evandro M. Lanzoni
- Brazilian
Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP 13083-970, Brazil
- Institute
of Science and Technology, São Paulo
State University (UNESP), Sorocaba, SP 18087-180, Brazil
| | - Carlos A. R. Costa
- Brazilian
Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP 13083-970, Brazil
| | - Paul M. Murray
- Paul
Murray Catalysis Consulting Ltd., 67 Hudson Close, Yate BS37 4NP, U.K.
| | - Christoph Deneke
- Brazilian
Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP 13083-970, Brazil
- Departamento
de Física Aplicada, Instituto de Física “Gleb
Wataghin”, Universidade Estadual
de Campinas − UNICAMP, Campinas, SP 13083-859, Brazil
| | - Fernando Galembeck
- Brazilian
Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP 13083-970, Brazil
| | - Janet L. Scott
- Department
of Chemical Engineering, Centre for Sustainable Chemical
Technologies, and Department of Chemistry, University of
Bath, Bath BA2 7AY, U.K.
| | - Ram I. Sharma
- Department
of Chemical Engineering, Centre for Sustainable Chemical
Technologies, and Department of Chemistry, University of
Bath, Bath BA2 7AY, U.K.
| |
Collapse
|
29
|
Abstract
In their native environment, cells are immersed in a complex milieu of biochemical and biophysical cues. These cues may include growth factors, the extracellular matrix, cell-cell contacts, stiffness, and topography, and they are responsible for regulating cellular behaviors such as adhesion, proliferation, migration, apoptosis, and differentiation. The decision-making process used to convert these extracellular inputs into actions is highly complex and sensitive to changes both in the type of individual cue (e.g., growth factor dose/level, timing) and in how these individual cues are combined (e.g., homotypic/heterotypic combinations). In this review, we highlight recent advances in the development of engineering-based approaches to study the cellular decision-making process. Specifically, we discuss the use of biomaterial platforms that enable controlled and tailored delivery of individual and combined cues, as well as the application of computational modeling to analyses of the complex cellular decision-making networks.
Collapse
Affiliation(s)
- Pamela K Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA; , .,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin 53705, USA.,Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705, USA.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| | - Laura E Strong
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA; ,
| | - Kristyn S Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA; , .,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA.,Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792, USA
| |
Collapse
|
30
|
Oswald J, Baranov P. Regenerative medicine in the retina: from stem cells to cell replacement therapy. Ther Adv Ophthalmol 2018; 10:2515841418774433. [PMID: 29998222 PMCID: PMC6016968 DOI: 10.1177/2515841418774433] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/15/2018] [Indexed: 12/20/2022] Open
Abstract
Following the fast pace of the growing field of stem cell research, retinal cell replacement is finally emerging as a feasible mean to be explored for clinical application. Although neuroprotective treatments are able to slow the progression of retinal degeneration caused by diseases such as age-related macular degeneration and glaucoma, they are insufficient to fully halt disease progression and unable to recover previously lost vision. Comprehensive, technological and intellectual advances over the past years, including the in vitro differentiation of retinal cells at manufacturing scale from embryonic stem (ES) cell and induced pluripotent stem (iPS) cell cultures, progress within the area of retinal disease modeling, and the first clinical trials have started to shape the way towards addressing this treatment gap and translating retinal cell replacement to the clinic. Here, summarize the most recent advances within retinal cell replacement from both a scientific and clinical perspective, and discuss the remaining challenges towards the delivery of the first retinal cell products.
Collapse
Affiliation(s)
- Julia Oswald
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, MA 02114, USA
| | - Petr Baranov
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, MA, USA
| |
Collapse
|
31
|
Toms D, Deardon R, Ungrin M. Climbing the mountain: experimental design for the efficient optimization of stem cell bioprocessing. J Biol Eng 2017; 11:35. [PMID: 29213303 PMCID: PMC5712411 DOI: 10.1186/s13036-017-0078-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/27/2017] [Indexed: 12/26/2022] Open
Abstract
"To consult the statistician after an experiment is finished is often merely to ask him to conduct a post mortem examination. He can perhaps say what the experiment died of." - R.A. Fisher While this idea is relevant across research scales, its importance becomes critical when dealing with the inherently large, complex and expensive process of preparing material for cell-based therapies (CBTs). Effective and economically viable CBTs will depend on the establishment of optimized protocols for the production of the necessary cell types. Our ability to do this will depend in turn on the capacity to efficiently search through a multi-dimensional problem space of possible protocols in a timely and cost-effective manner. In this review we discuss approaches to, and illustrate examples of the application of statistical design of experiments to stem cell bioprocess optimization.
Collapse
Affiliation(s)
- Derek Toms
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, T2N 4Z6 Canada
| | - Rob Deardon
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, T2N 4Z6 Canada
- Department of Mathematics and Statistics, Faculty of Science, University of Calgary, 612 Campus Place NW, Calgary, T2N 4N1 Canada
| | - Mark Ungrin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, T2N 4Z6 Canada
- Biomedical Engineering Graduate Program, University of Calgary, 2500 University Drive NW, Calgary, T2N 1N4 Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1 Canada
- Alberta Diabetes Institute, University of Alberta, Li Ka Shing Centre for Health Research Innovation, Edmonton, T6G 2E1 Canada
- Centre for Bioengineering Research and Education, University of Calgary, 2500 University Drive NW, Calgary, T2N 1N4 Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, 3280 Hospital Drive NW, Calgary, T2N 4Z6 Canada
| |
Collapse
|
32
|
Mora-Solano C, Wen Y, Han H, Chen J, Chong AS, Miller ML, Pompano RR, Collier JH. Active immunotherapy for TNF-mediated inflammation using self-assembled peptide nanofibers. Biomaterials 2017; 149:1-11. [PMID: 28982051 DOI: 10.1016/j.biomaterials.2017.09.031] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 11/18/2022]
Abstract
Active immunotherapies raising antibody responses against autologous targets are receiving increasing interest as alternatives to the administration of manufactured antibodies. The challenge in such an approach is generating protective and adjustable levels of therapeutic antibodies while at the same time avoiding strong T cell responses that could lead to autoimmune reactions. Here we demonstrate the design of an active immunotherapy against TNF-mediated inflammation using short synthetic peptides that assemble into supramolecular peptide nanofibers. Immunization with these materials, without additional adjuvants, was able to break B cell tolerance and raise protective antibody responses against autologous TNF in mice. The strength of the anti-TNF antibody response could be tuned by adjusting the epitope content in the nanofibers, and the T-cell response was focused on exogenous and non-autoreactive T-cell epitopes. Immunization with unadjuvanted peptide nanofibers was therapeutic in a lethal model of acute inflammation induced by intraperitoneally delivered lipopolysaccharide, whereas formulations adjuvanted with CpG showed comparatively poorer protection that correlated with a more Th1-polarized response. Additionally, immunization with peptide nanofibers did not diminish the ability of mice to clear infections of Listeria monocytogenes. Collectively this work suggests that synthetic self-assembled peptides can be attractive platforms for active immunotherapies against autologous targets.
Collapse
Affiliation(s)
- Carolina Mora-Solano
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States; Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States
| | - Yi Wen
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Huifang Han
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Jianjun Chen
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Anita S Chong
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Michelle L Miller
- Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22904, United States
| | - Joel H Collier
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States; Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States.
| |
Collapse
|
33
|
Abstract
The development of biomaterials designed for specific applications is an important objective in personalized medicine. While the breadth and prominence of biomaterials have increased exponentially over the past decades, critical challenges remain to be addressed, particularly in the development of biomaterials that exhibit highly specific functions. These functional properties are often encoded within the molecular structure of the component molecules. Proteins, as a consequence of their structural specificity, represent useful substrates for the construction of functional biomaterials through rational design. This chapter provides an in-depth survey of biomaterials constructed from coiled-coils, one of the best-understood protein structural motifs. We discuss the utility of this structurally diverse and functionally tunable class of proteins for the creation of novel biomaterials. This discussion illustrates the progress that has been made in the development of coiled-coil biomaterials by showcasing studies that bridge the gap between the academic science and potential technological impact.
Collapse
Affiliation(s)
- David A.D. Parry
- Institute of Fundamental Sciences and Riddet Institute, Massey University, Palmerston North, New Zealand
| | - John M. Squire
- Muscle Contraction Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
34
|
Clarke D, Pashuck ET, Bertazzo S, Weaver JVM, Stevens MM. Self-Healing, Self-Assembled β-Sheet Peptide-Poly(γ-glutamic acid) Hybrid Hydrogels. J Am Chem Soc 2017; 139:7250-7255. [PMID: 28525280 PMCID: PMC5467180 DOI: 10.1021/jacs.7b00528] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Indexed: 12/29/2022]
Abstract
Self-assembled biomaterials are an important class of materials that can be injected and formed in situ. However, they often are not able to meet the mechanical properties necessary for many biological applications, losing mechanical properties at low strains. We synthesized hybrid hydrogels consisting of a poly(γ-glutamic acid) polymer network physically cross-linked via grafted self-assembling β-sheet peptides to provide non-covalent cross-linking through β-sheet assembly, reinforced with a polymer backbone to improve strain stability. By altering the β-sheet peptide graft density and concentration, we can tailor the mechanical properties of the hydrogels over an order of magnitude range of 10-200 kPa, which is in the region of many soft tissues. Also, due to the ability of the non-covalent β-sheet cross-links to reassemble, the hydrogels can self-heal after being strained to failure, in most cases recovering all of their original storage moduli. Using a combination of spectroscopic techniques, we were able to probe the secondary structure of the materials and verify the presence of β-sheets within the hybrid hydrogels. Since the polymer backbone requires less than a 15% functionalization of its repeating units with β-sheet peptides to form a hydrogel, it can easily be modified further to incorporate specific biological epitopes. This self-healing polymer-β-sheet peptide hybrid hydrogel with tailorable mechanical properties is a promising platform for future tissue-engineering scaffolds and biomedical applications.
Collapse
Affiliation(s)
- David
E. Clarke
- Department
of Materials, Institute of Biomedical Engineering,
and Department of Bioengineering, Imperial College London, Exhibition Road, London, SW7 2AZ, U.K.
| | - E. Thomas Pashuck
- Department
of Materials, Institute of Biomedical Engineering,
and Department of Bioengineering, Imperial College London, Exhibition Road, London, SW7 2AZ, U.K.
| | - Sergio Bertazzo
- Department
of Materials, Institute of Biomedical Engineering,
and Department of Bioengineering, Imperial College London, Exhibition Road, London, SW7 2AZ, U.K.
| | - Jonathan V. M. Weaver
- Department
of Materials, Institute of Biomedical Engineering,
and Department of Bioengineering, Imperial College London, Exhibition Road, London, SW7 2AZ, U.K.
| | - Molly M. Stevens
- Department
of Materials, Institute of Biomedical Engineering,
and Department of Bioengineering, Imperial College London, Exhibition Road, London, SW7 2AZ, U.K.
| |
Collapse
|
35
|
Lim HJ, Khan Z, Wilems TS, Lu X, Perera TH, Kurosu YE, Ravivarapu KT, Mosley MC, Smith Callahan LA. Human Induced Pluripotent Stem Cell Derived Neural Stem Cell Survival and Neural Differentiation on Polyethylene Glycol Dimethacrylate Hydrogels Containing a Continuous Concentration Gradient of N-Cadherin Derived Peptide His-Ala-Val-Asp-Ile. ACS Biomater Sci Eng 2017; 3:776-781. [DOI: 10.1021/acsbiomaterials.6b00745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Hyun Ju Lim
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Zara Khan
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Thomas S. Wilems
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Xi Lu
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - T. Hiran Perera
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Yuki E. Kurosu
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Krishna T. Ravivarapu
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Matthew C. Mosley
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| | - Laura A. Smith Callahan
- The
Vivian L. Smith Department of Neurosurgery and Center for Stem Cells
and Regenerative Medicine, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
- Department
of Nanomedicine and Biomedical Engineering, McGovern Medical School at the University of Texas Health Science Center at Houston, 1825 Pressler, Houston, Texas 77030, United States
| |
Collapse
|
36
|
Heath DE, Cooper SL. The development of polymeric biomaterials inspired by the extracellular matrix. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1051-1069. [DOI: 10.1080/09205063.2017.1297285] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Daniel E. Heath
- Department of Chemical and Biomolecular Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Parkville, Australia
| | - Stuart L. Cooper
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
37
|
Affiliation(s)
- Daniel E. Heath
- Department of Chemical and Biomolecular Engineering; Particulate Fluids Processing Centre; The University of Melbourne; Parkville Victoria Australia
| |
Collapse
|
38
|
Yu Z, Cai Z, Chen Q, Liu M, Ye L, Ren J, Liao W, Liu S. Engineering β-sheet peptide assemblies for biomedical applications. Biomater Sci 2017; 4:365-74. [PMID: 26700207 DOI: 10.1039/c5bm00472a] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hydrogels have been widely studied in various biomedical applications, such as tissue engineering, cell culture, immunotherapy and vaccines, and drug delivery. Peptide-based nanofibers represent a promising new strategy for current drug delivery approaches and cell carriers for tissue engineering. This review focuses on the recent advances in the use of self-assembling engineered β-sheet peptide assemblies for biomedical applications. The applications of peptide nanofibers in biomedical fields, such as drug delivery, tissue engineering, immunotherapy, and vaccines, are highlighted. The current challenges and future perspectives for self-assembling peptide nanofibers in biomedical applications are discussed.
Collapse
Affiliation(s)
- Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| | - Zheng Cai
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| | - Qiling Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| | - Menghua Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| | - Ling Ye
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| | - Jiaoyan Ren
- Department of Food Science and Technology, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong, China.
| | - Wenzhen Liao
- Department of Food Science and Technology, South China University of Technology, Wushan Road 381, Guangzhou, Guangdong, China.
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
39
|
Pagel M, Beck-Sickinger AG. Multifunctional biomaterial coatings: synthetic challenges and biological activity. Biol Chem 2017; 398:3-22. [DOI: 10.1515/hsz-2016-0204] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/29/2016] [Indexed: 12/19/2022]
Abstract
Abstract
A controlled interaction of materials with their surrounding biological environment is of great interest in many fields. Multifunctional coatings aim to provide simultaneous modulation of several biological signals. They can consist of various combinations of bioactive, and bioinert components as well as of reporter molecules to improve cell-material contacts, prevent infections or to analyze biochemical events on the surface. However, specific immobilization and particular assembly of various active molecules are challenging. Herein, an overview of multifunctional coatings for biomaterials is given, focusing on synthetic strategies and the biological benefits by displaying several motifs.
Collapse
|
40
|
Co-Assembly Tags Based on Charge Complementarity (CATCH) for Installing Functional Protein Ligands into Supramolecular Biomaterials. Cell Mol Bioeng 2016. [DOI: 10.1007/s12195-016-0459-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
41
|
Smith Callahan LA. Combinatorial Method/High Throughput Strategies for Hydrogel Optimization in Tissue Engineering Applications. Gels 2016; 2:E18. [PMID: 30674150 PMCID: PMC6318679 DOI: 10.3390/gels2020018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/01/2016] [Accepted: 06/03/2016] [Indexed: 12/22/2022] Open
Abstract
Combinatorial method/high throughput strategies, which have long been used in the pharmaceutical industry, have recently been applied to hydrogel optimization for tissue engineering applications. Although many combinatorial methods have been developed, few are suitable for use in tissue engineering hydrogel optimization. Currently, only three approaches (design of experiment, arrays and continuous gradients) have been utilized. This review highlights recent work with each approach. The benefits and disadvantages of design of experiment, array and continuous gradient approaches depending on study objectives and the general advantages of using combinatorial methods for hydrogel optimization over traditional optimization strategies will be discussed. Fabrication considerations for combinatorial method/high throughput samples will additionally be addressed to provide an assessment of the current state of the field, and potential future contributions to expedited material optimization and design.
Collapse
Affiliation(s)
- Laura A Smith Callahan
- Vivian L. Smith Department of Neurosurgery & Center for Stem Cells and Regenerative Medicine McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
- Department of Nanomedicine and Biomedical Engineering, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Vigneswaran Y, Han H, De Loera R, Wen Y, Zhang X, Sun T, Mora-Solano C, Collier JH. This paper is the winner of an SFB Award in the Hospital Intern, Residency category: Peptide biomaterials raising adaptive immune responses in wound healing contexts. J Biomed Mater Res A 2016; 104:1853-62. [PMID: 27129604 DOI: 10.1002/jbm.a.35767] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022]
Abstract
Biomaterials used in the context of tissue engineering or wound repair are commonly designed to be "nonimmunogenic." However, previously it has been observed that self-assembled peptide nanofiber materials are noninflammatory despite their immunogenicity, suggesting that they may be appropriate for use in wound-healing contexts. To test this hypothesis, mice were immunized with epitope-containing peptide self-assemblies until they maintained high antibody titers against the material, then gels of the same peptide assemblies were applied within full-thickness dermal wounds. In three different murine dermal-wounding models with different baseline healing rates, even significantly immunogenic peptide assemblies did not delay healing. Conversely, adjuvanted peptide assemblies, while raising similar antibody titers to unadjuvanted assemblies, did delay wound healing. Analysis of the healing wounds indicated that compared to adjuvanted peptide assemblies, the unadjuvanted assemblies exhibited a progression of the dominant T-cell subset from CD4(+) to CD8(+) cells in the wound, and CD4(+) cell populations displayed a more Th2-slanted response. These findings illustrate an example of a significant antibiomaterial adaptive immune response that does not adversely affect wound healing despite ongoing antibody production. This material would thus be considered "immunologically compatible" in this specific context rather than "nonimmunogenic," a designation that is expected to apply to a range of other protein- and peptide-based biomaterials in wound-healing and tissue-engineering applications. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1853-1862, 2016.
Collapse
Affiliation(s)
| | - Huifang Han
- Department of Surgery, University of Chicago, Chicago, Illinois
| | | | - Yi Wen
- Department of Surgery, University of Chicago, Chicago, Illinois.,Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Xing Zhang
- Department of Surgery, University of Chicago, Chicago, Illinois
| | - Tao Sun
- Department of Surgery, University of Chicago, Chicago, Illinois
| | | | - Joel H Collier
- Department of Surgery, University of Chicago, Chicago, Illinois.,Department of Biomedical Engineering, Duke University, Durham, North Carolina
| |
Collapse
|
43
|
Elsawy M, Smith AM, Hodson N, Squires A, Miller AF, Saiani A. Modification of β-Sheet Forming Peptide Hydrophobic Face: Effect on Self-Assembly and Gelation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:4917-23. [PMID: 27089379 PMCID: PMC4990315 DOI: 10.1021/acs.langmuir.5b03841] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
β-Sheet forming peptides have attracted significant interest for the design of hydrogels for biomedical applications. One of the main challenges is the control and understanding of the correlations between peptide molecular structure, the morphology, and topology of the fiber and network formed as well as the macroscopic properties of the hydrogel obtained. In this work, we have investigated the effect that functionalizing these peptides through their hydrophobic face has on their self-assembly and gelation. Our results show that the modification of the hydrophobic face results in a partial loss of the extended β-sheet conformation of the peptide and a significant change in fiber morphology from straight to kinked. As a consequence, the ability of these fibers to associate along their length and form large bundles is reduced. These structural changes (fiber structure and network topology) significantly affect the mechanical properties of the hydrogels (shear modulus and elasticity).
Collapse
Affiliation(s)
- Mohamed
A. Elsawy
- School of Materials, Manchester Institute of Biotechnology, BioAFM Facility, Stopford
Building, and School of Chemical Engineering and Analytical Science, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
| | - Andrew M. Smith
- School of Materials, Manchester Institute of Biotechnology, BioAFM Facility, Stopford
Building, and School of Chemical Engineering and Analytical Science, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
| | - Nigel Hodson
- School of Materials, Manchester Institute of Biotechnology, BioAFM Facility, Stopford
Building, and School of Chemical Engineering and Analytical Science, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
| | - Adam Squires
- Department
of Chemistry, Reading University, Whiteknights RG6 6AD, Reading, U.K.
| | - Aline F. Miller
- School of Materials, Manchester Institute of Biotechnology, BioAFM Facility, Stopford
Building, and School of Chemical Engineering and Analytical Science, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
| | - Alberto Saiani
- School of Materials, Manchester Institute of Biotechnology, BioAFM Facility, Stopford
Building, and School of Chemical Engineering and Analytical Science, The University of Manchester, Oxford Road, M13
9PL Manchester, U.K.
- Phone +44
161 306 5981; Fax +44 161 306 3586; e-mail (A.S.)
| |
Collapse
|
44
|
Rehmann MS, Luna JI, Maverakis E, Kloxin AM. Tuning microenvironment modulus and biochemical composition promotes human mesenchymal stem cell tenogenic differentiation. J Biomed Mater Res A 2016; 104:1162-74. [PMID: 26748903 PMCID: PMC5510610 DOI: 10.1002/jbm.a.35650] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/19/2015] [Accepted: 01/08/2016] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are promising for the regeneration of tendon and ligament tissues. Toward realizing this potential, microenvironment conditions are needed for promoting robust lineage-specific differentiation into tenocytes/ligament fibroblasts. Here, we utilized a statistical design of experiments approach to examine combinations of matrix modulus, composition, and soluble factors in human MSC tenogenic/ligamentogenic differentiation. Specifically, well-defined poly(ethylene glycol)-based hydrogels were synthesized using thiol-ene chemistry providing a bioinert base for probing cell response to extracellular matrix cues. Monomer concentrations were varied to achieve a range of matrix moduli (E ∼ 10-90 kPa), and different ratios of integrin-binding peptides were incorporated (GFOGER and RGDS for collagen and fibronectin, respectively), mimicking aspects of developing tendon/ligament tissue. A face-centered central composite response surface design was utilized to understand the contributions of these cues to human MSC differentiation in the presence of soluble factors identified to promote tenogenesis/ligamentogenesis (BMP-13 and ascorbic acid). Increasing modulus and collagen mimetic peptide content increased relevant gene expression and protein production or retention (scleraxis, collagen I, tenascin-C). These findings could inform the design of materials for tendon/ligament regeneration. More broadly, the design of experiments enabled efficient data acquisition and analysis, requiring fewer replicates than if each factor had been varied one at a time. This approach can be combined with other stimuli (for example, mechanical stimulation) toward a better mechanistic understanding of differentiation down these challenging lineages.
Collapse
Affiliation(s)
- Matthew S Rehmann
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, 19716
| | - Jesus I Luna
- Department of Dermatology, School of Medicine, University of California, Davis, California, 95816
| | - Emanual Maverakis
- Department of Dermatology, School of Medicine, University of California, Davis, California, 95816
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, 19716
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716
| |
Collapse
|
45
|
Huber B, Engelhardt S, Meyer W, Krüger H, Wenz A, Schönhaar V, Tovar GEM, Kluger PJ, Borchers K. Blood-Vessel Mimicking Structures by Stereolithographic Fabrication of Small Porous Tubes Using Cytocompatible Polyacrylate Elastomers, Biofunctionalization and Endothelialization. J Funct Biomater 2016; 7:E11. [PMID: 27104576 PMCID: PMC4932468 DOI: 10.3390/jfb7020011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/18/2016] [Accepted: 04/08/2016] [Indexed: 12/16/2022] Open
Abstract
Blood vessel reconstruction is still an elusive goal for the development of in vitro models as well as artificial vascular grafts. In this study, we used a novel photo-curable cytocompatible polyacrylate material (PA) for freeform generation of synthetic vessels. We applied stereolithography for the fabrication of arbitrary 3D tubular structures with total dimensions in the centimeter range, 300 µm wall thickness, inner diameters of 1 to 2 mm and defined pores with a constant diameter of approximately 100 µm or 200 µm. We established a rinsing protocol to remove remaining cytotoxic substances from the photo-cured PA and applied thio-modified heparin and RGDC-peptides to functionalize the PA surface for enhanced endothelial cell adhesion. A rotating seeding procedure was introduced to ensure homogenous endothelial monolayer formation at the inner luminal tube wall. We showed that endothelial cells stayed viable and adherent and aligned along the medium flow under fluid-flow conditions comparable to native capillaries. The combined technology approach comprising of freeform additive manufacturing (AM), biomimetic design, cytocompatible materials which are applicable to AM, and biofunctionalization of AM constructs has been introduced as BioRap(®) technology by the authors.
Collapse
Affiliation(s)
- Birgit Huber
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart 70569, Germany.
| | - Sascha Engelhardt
- Rheinisch-Westfälische Technische Hochschule Aachen, RWTH Aachen, Aachen 52074, Germany.
| | - Wolfdietrich Meyer
- Fraunhofer Institute for Applied Polymer Research IAP, Potsdam 14476, Germany.
| | - Hartmut Krüger
- Fraunhofer Institute for Applied Polymer Research IAP, Potsdam 14476, Germany.
| | - Annika Wenz
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart 70569, Germany.
| | - Veronika Schönhaar
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany.
| | - Günter E M Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart 70569, Germany.
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany.
| | - Petra J Kluger
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany.
- Process Analysis & Technology (PA&T), Reutlingen University, Reutlingen 72762, Germany.
| | - Kirsten Borchers
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart 70569, Germany.
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart 70569, Germany.
| |
Collapse
|
46
|
Liu W, Saunders MJ, Bagia C, Freeman EC, Fan Y, Gawalt ES, Waggoner AS, Meng WS. Local retention of antibodies in vivo with an injectable film embedded with a fluorogen-activating protein. J Control Release 2016; 230:1-12. [PMID: 27038493 DOI: 10.1016/j.jconrel.2016.03.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 02/25/2016] [Accepted: 03/22/2016] [Indexed: 11/17/2022]
Abstract
Herein we report an injectable film by which antibodies can be localized in vivo. The system builds upon a bifunctional polypeptide consisting of a fluorogen-activating protein (FAP) and a β-fibrillizing peptide (βFP). The FAP domain generates fluorescence that reflects IgG binding sites conferred by Protein A/G (pAG) conjugated with the fluorogen malachite green (MG). A film is generated by mixing these proteins with molar excess of EAK16-II, a βFP that forms β-sheet fibrils at high salt concentrations. The IgG-binding, fluorogenic film can be injected in vivo through conventional needled syringes. Confocal microscopic images and dose-response titration experiments showed that loading of IgG into the film was mediated by pAG(MG) bound to the FAP. Release of IgG in vitro was significantly delayed by the bioaffinity mechanism; 26% of the IgG were released from films embedded with pAG(MG) after five days, compared to close to 90% in films without pAG(MG). Computational simulations indicated that the release rate of IgG is governed by positive cooperativity due to pAG(MG). When injected into the subcutaneous space of mouse footpads, film-embedded IgG were retained locally, with distribution through the lymphatics impeded. The ability to track IgG binding sites and distribution simultaneously will aid the optimization of local antibody delivery systems.
Collapse
Affiliation(s)
- Wen Liu
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States
| | - Matthew J Saunders
- Molecular Biosensor and Imaging Center and Carnegie Mellon University, Pittsburgh, PA 15213, United States; Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Christina Bagia
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States
| | - Eric C Freeman
- College of Engineering, University of Georgia, Athens, GA 30602, United States
| | - Yong Fan
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Pittsburgh, PA 15212, United States; Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Ellen S Gawalt
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, PA 15282, United States; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15213, United States
| | - Alan S Waggoner
- Molecular Biosensor and Imaging Center and Carnegie Mellon University, Pittsburgh, PA 15213, United States; Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Wilson S Meng
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States.
| |
Collapse
|
47
|
Jung JP, Hu D, Domian IJ, Ogle BM. An integrated statistical model for enhanced murine cardiomyocyte differentiation via optimized engagement of 3D extracellular matrices. Sci Rep 2015; 5:18705. [PMID: 26687770 PMCID: PMC4685314 DOI: 10.1038/srep18705] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/24/2015] [Indexed: 01/28/2023] Open
Abstract
The extracellular matrix (ECM) impacts stem cell differentiation, but identifying formulations supportive of differentiation is challenging in 3D models. Prior efforts involving combinatorial ECM arrays seemed intuitively advantageous. We propose an alternative that suggests reducing sample size and technological burden can be beneficial and accessible when coupled to design of experiments approaches. We predict optimized ECM formulations could augment differentiation of cardiomyocytes derived in vitro. We employed native chemical ligation to polymerize 3D poly (ethylene glycol) hydrogels under mild conditions while entrapping various combinations of ECM and murine induced pluripotent stem cells. Systematic optimization for cardiomyocyte differentiation yielded a predicted solution of 61%, 24%, and 15% of collagen type I, laminin-111, and fibronectin, respectively. This solution was confirmed by increased numbers of cardiac troponin T, α-myosin heavy chain and α-sarcomeric actinin-expressing cells relative to suboptimum solutions. Cardiomyocytes of composites exhibited connexin43 expression, appropriate contractile kinetics and intracellular calcium handling. Further, adding a modulator of adhesion, thrombospondin-1, abrogated cardiomyocyte differentiation. Thus, the integrated biomaterial platform statistically identified an ECM formulation best supportive of cardiomyocyte differentiation. In future, this formulation could be coupled with biochemical stimulation to improve functional maturation of cardiomyocytes derived in vitro or transplanted in vivo.
Collapse
Affiliation(s)
- Jangwook P Jung
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A.,Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A
| | - Dongjian Hu
- Cardiovascular Research Center, Massachusetts General Hospital &Harvard Medical School, Boston, MA 02114 U.S.A
| | - Ibrahim J Domian
- Cardiovascular Research Center, Massachusetts General Hospital &Harvard Medical School, Boston, MA 02114 U.S.A
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A.,Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A.,Masonic Cancer Center, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A.,Lillehei Heart Institute, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A.,Institute for Engineering in Medicine, University of Minnesota - Twin Cities, Minneapolis, MN 55455, U.S.A
| |
Collapse
|
48
|
Rad-Malekshahi M, Lempsink L, Amidi M, Hennink WE, Mastrobattista E. Biomedical Applications of Self-Assembling Peptides. Bioconjug Chem 2015; 27:3-18. [DOI: 10.1021/acs.bioconjchem.5b00487] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mazda Rad-Malekshahi
- Department of Pharmaceutics,
Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584
CG Utrecht, The Netherlands
| | - Ludwijn Lempsink
- Department of Pharmaceutics,
Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584
CG Utrecht, The Netherlands
| | - Maryam Amidi
- Department of Pharmaceutics,
Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584
CG Utrecht, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics,
Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584
CG Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department of Pharmaceutics,
Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584
CG Utrecht, The Netherlands
| |
Collapse
|
49
|
Loo Y, Goktas M, Tekinay AB, Guler MO, Hauser CAE, Mitraki A. Self-Assembled Proteins and Peptides as Scaffolds for Tissue Regeneration. Adv Healthc Mater 2015; 4:2557-86. [PMID: 26461979 DOI: 10.1002/adhm.201500402] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/24/2015] [Indexed: 12/15/2022]
Abstract
Self-assembling proteins and peptides are increasingly gaining interest for potential use as scaffolds in tissue engineering applications. They self-organize from basic building blocks under mild conditions into supramolecular structures, mimicking the native extracellular matrix. Their properties can be easily tuned through changes at the sequence level. Moreover, they can be produced in sufficient quantities with chemical synthesis or recombinant technologies to allow them to address homogeneity and standardization issues required for applications. Here. recent advances in self-assembling proteins, peptides, and peptide amphiphiles that form scaffolds suitable for tissue engineering are reviewed. The focus is on a variety of motifs, ranging from minimalistic dipeptides, simplistic ultrashort aliphatic peptides, and peptide amphiphiles to large "recombinamer" proteins. Special emphasis is placed on the rational design of self-assembling motifs and biofunctionalization strategies to influence cell behavior and modulate scaffold stability. Perspectives for combination of these "bottom-up" designer strategies with traditional "top-down" biofabrication techniques for new generations of tissue engineering scaffolds are highlighted.
Collapse
Affiliation(s)
- Yihua Loo
- Institute for Bioengineering and Nanotechnology; A* STAR; 31 Biopolis Way The Nanos 138669 Singapore
| | - Melis Goktas
- Institute of Materials Science and Nanotechnology; National Nanotechnology Research Center (UNAM); Bilkent University; Ankara Turkey 06800
| | - Ayse B. Tekinay
- Institute of Materials Science and Nanotechnology; National Nanotechnology Research Center (UNAM); Bilkent University; Ankara Turkey 06800
| | - Mustafa O. Guler
- Institute of Materials Science and Nanotechnology; National Nanotechnology Research Center (UNAM); Bilkent University; Ankara Turkey 06800
| | - Charlotte A. E. Hauser
- Institute for Bioengineering and Nanotechnology; A* STAR; 31 Biopolis Way The Nanos 138669 Singapore
| | - Anna Mitraki
- Department of Materials Science and Technology; University of Crete; Greece 70013
- Institute for Electronic Structure and Lasers (IESL); Foundation for Research and Technology Hellas (FORTH); Vassilika Vouton; Heraklion Crete Greece 70013
| |
Collapse
|
50
|
Abstract
Strategies to enhance, suppress, or qualitatively shape the immune response are of importance for diverse biomedical applications, such as the development of new vaccines, treatments for autoimmune diseases and allergies, strategies for regenerative medicine, and immunotherapies for cancer. However, the intricate cellular and molecular signals regulating the immune system are major hurdles to predictably manipulating the immune response and developing safe and effective therapies. To meet this challenge, biomaterials are being developed that control how, where, and when immune cells are stimulated in vivo, and that can finely control their differentiation in vitro. We review recent advances in the field of biomaterials for immunomodulation, focusing particularly on designing biomaterials to provide controlled immunostimulation, targeting drugs and vaccines to lymphoid organs, and serving as scaffolds to organize immune cells and emulate lymphoid tissues. These ongoing efforts highlight the many ways in which biomaterials can be brought to bear to engineer the immune system.
Collapse
Affiliation(s)
- Nathan A Hotaling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
| | - Li Tang
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
| | - Darrell J Irvine
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| | - Julia E Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia 30332;
| |
Collapse
|