1
|
Hussain S, Maktedar SS. Structural, functional and mechanical performance of advanced Graphene-based composite hydrogels. RESULTS IN CHEMISTRY 2023; 6:101029. [DOI: 10.1016/j.rechem.2023.101029] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
2
|
Gonzalez-Valdivieso J, Vallejo R, Rodriguez-Rojo S, Santos M, Schneider J, Arias FJ, Girotti A. CD44-targeted nanoparticles for co-delivery of docetaxel and an Akt inhibitor against colorectal cancer. BIOMATERIALS ADVANCES 2023; 154:213595. [PMID: 37639856 DOI: 10.1016/j.bioadv.2023.213595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/24/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
New strategies to develop drug-loaded nanocarriers with improved therapeutic efficacy are needed for cancer treatment. Herein we report a novel drug-delivery nanosystem comprising encapsulation of the chemotherapeutic drug docetaxel (DTX) and recombinant fusion of a small peptide inhibitor of Akt kinase within an elastin-like recombinamer (ELR) vehicle. This combined approach is also precisely targeted to colorectal cancer cells by means of a chemically conjugated DNA aptamer specific for the CD44 tumor marker. This 53 nm dual-approach nanosystem was found to selectively affect cell viability (2.5 % survival) and proliferation of colorectal cancer cells in vitro compared to endothelial cells (50 % survival), and to trigger both apoptosis- and necrosis-mediated cell death. Our findings also show that the nanohybrid particles remain stable under physiological conditions, trigger sustained drug release and possess an adequate pharmacokinetic profile after systemic intravenous administration. In vivo assays showed that these dual-approach nanohybrids significantly reduced the number of tumor polyps along the colorectal tract in a murine colorectal cancer model. Furthermore, systemic administration of advanced nanohybrids induced tissue recovery by improving the morphology of gastrointestinal crypts and the tissue architecture. Taken together, these findings indicate that our strategy of an advanced dual-approach nanosystem allows us to achieve successful controlled release of chemotherapeutics in cancer cells and may have a promising potential for colorectal cancer treatment.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain
| | - Reinaldo Vallejo
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Soraya Rodriguez-Rojo
- BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Mercedes Santos
- BIOFORGE Research Group (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Valladolid, Spain
| | - Jose Schneider
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Department of Obstetrics & Gynecology, University of Valladolid, School of Medicine, Valladolid, Spain
| | - Francisco Javier Arias
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| | - Alessandra Girotti
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| |
Collapse
|
3
|
Gonzalez-Valdivieso J, Garcia-Sampedro A, Hall AR, Girotti A, Arias FJ, Pereira SP, Acedo P. Smart Nanoparticles as Advanced Anti-Akt Kinase Delivery Systems for Pancreatic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:55790-55805. [PMID: 34788541 DOI: 10.1021/acsami.1c14592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pancreatic cancer is one of the deadliest cancers partly due to late diagnosis, poor drug delivery to the target site, and acquired resistance to therapy. Therefore, more effective therapies are urgently needed to improve the outcome of patients. In this work, we have tested self-assembling genetically engineered polymeric nanoparticles formed by elastin-like recombinamers (ELRs), carrying a small peptide inhibitor of the protein kinase Akt, in both PANC-1 and patient-derived pancreatic cancer cells (PDX models). Nanoparticle cell uptake was measured by flow cytometry, and subcellular localization was determined by confocal microscopy, which showed a lysosomal localization of these nanoparticles. Furthermore, metabolic activity and cell viability were significantly reduced after incubation with nanoparticles carrying the Akt inhibitor in a time- and dose-dependent fashion. Self-assembling 73 ± 3.2 nm size nanoparticles inhibited phosphorylation and consequent activation of Akt protein, blocked the NF-κB signaling pathway, and triggered caspase 3-mediated apoptosis. Furthermore, in vivo assays showed that ELR-based nanoparticles were suitable devices for drug delivery purposes with long circulating time and minimum toxicity. Hence, the use of these smart nanoparticles could lead to the development of more effective treatment options for pancreatic cancer based on the inhibition of Akt.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Biodevices for NanoMed Group, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Andres Garcia-Sampedro
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Andrew R Hall
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, United Kingdom
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Francisco Javier Arias
- Smart Biodevices for NanoMed Group, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| |
Collapse
|
4
|
Chiang MY, Lo YC, Lai YH, Yong YYA, Chang SJ, Chen WL, Chen SY. Protein-based soft actuator with high photo-response and easy modulation for anisotropic cell alignment and proliferation in a liquid environment. J Mater Chem B 2021; 9:6634-6645. [PMID: 34365493 DOI: 10.1039/d1tb01198g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cell alignment and elongation, which are critical factors correlated with differentiation and maturation in cell biology and tissue engineering, have been widely studied in organisms. Several strategies such as external mechanical strain, geometric topography, micropatterning approaches, and microfabricated substrates have been developed to guide cell alignment, but these methodologies cannot be used for easily denatured natural proteins to modulate the cell behaviour. Herein, for the first time, a novel biocompatible light-controlled protein-based bilayer soft actuator composed of elastin-like polypeptides (ELPs), silk fibroin (SF), graphene oxide (GO), and reduced graphene oxide (rGO), named ESGRG, is developed for efficiently driving cellular orientation and elongation with anisotropic features on soft actuator via remote NIR laser exposure. The actuation of ESGRG could be manipulated by modulating the intensity of NIR and the relative ratio of GO to rGO for promoting myoblasts alignment and nucleus elongation to generate different motions. The results indicate that the YAP and MHC protein expression of C2C12 skeletal muscle cells on ESGRG can be rapidly induced and enhanced by controlling the relative ratio of rGO/GO = 1/4 at a multiple-cycle stimulation with a very low power intensity of 1.2 W cm-2 in friendly liquid environments. This study demonstrates that the ESGRG hydrogel actuator system can modulate the cell-level behaviors via light-driven cyclic bending-motions and can be utilized in applications of soft robotic and tissue engineering such as artificial muscle and maturation of cardiomyocytes.
Collapse
Affiliation(s)
- Min-Yu Chiang
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, No. 1001 Ta-Hsueh Rd, Hsinchu, Taiwan 300, Republic of China.
| | | | | | | | | | | | | |
Collapse
|
5
|
Torgbo S, Sukyai P. Biodegradation and thermal stability of bacterial cellulose as biomaterial: The relevance in biomedical applications. Polym Degrad Stab 2020. [DOI: 10.1016/j.polymdegradstab.2020.109232] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
6
|
Human plasma protein adsorption to elastin-like polypeptide nanoparticles. Biointerphases 2020; 15:021007. [DOI: 10.1116/6.0000027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
7
|
Lee KM, Kim JH, Choi ES, Kim E, Choi SK, Jeon WB. RGD-containing elastin-like polypeptide improves islet transplantation outcomes in diabetic mice. Acta Biomater 2019; 94:351-360. [PMID: 31200117 DOI: 10.1016/j.actbio.2019.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/24/2019] [Accepted: 06/10/2019] [Indexed: 12/22/2022]
Abstract
Successful islet transplantation critically depends on the isolation of healthy islets. However, the islet isolation procedure itself contributes to islet death due to the destruction of intra- and peri-islet extracellular matrices (ECMs) during digestion. We investigated whether an RGD-containing elastin-like polypeptide (REP) could function as a self-assembling matrix to replenish ECMs and protects islets from cell death. Immediately following isolation, islets were coated with REP coacervate particles via isothermal adsorption of an REP solution followed by thermal gelation. REP-coated islets displayed increased viability and insulin secretory capacity in pretransplant culture compared to untreated islets. Co-transplantation of REP-treated islets and REP beneath the renal sub-capsule in streptozotocin-induced diabetic mice restored normoglycemia and serum insulin levels. Mice that received co-transplants maintained normoglycemia for a longer period of time than those receiving untreated islets without REP. Moreover, co-transplantation sites exhibited enhanced β-cell proliferation and vascularization. Thus, the REP-based coacervation strategy improve the survival, function and therapeutic potential of transplanted islets. STATEMENT OF SIGNIFICANCE: 1). An artificial matrix polypeptide comprised of thermoresponsive elastin-like peptides and integrin-stimulatory RGD ligands (REP) to reconstitute damaged or lost matrices. 2). Through body temperature-induced coacervation, REP reconstitutes intra-islet environment and enhances islet viability and insulin secretion by activating the pro-survival and insulin signaling pathways. 3). REP-coated islets were transplanted together with the matrix polypeptide under the kidney sub-capsule of mice, it develops a new peri-insular environment, which protects the islet grafts from immune rejection thus extending islet longevity. 4). Our data suggest that in situ self-assembly of biomimetic islet environments become a new platform allowing for improved islet transplantation at extrahepatic sites.
Collapse
Affiliation(s)
- Kyeong-Min Lee
- Laboratory of Biochemistry and Cellular Engineering, Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Jung-Hee Kim
- Laboratory of Biochemistry and Cellular Engineering, Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Eun-Sook Choi
- Laboratory of Biochemistry and Cellular Engineering, Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Eunjoo Kim
- Laboratory of Biochemistry and Cellular Engineering, Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Seong-Kyoon Choi
- Core Protein Resources Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Won Bae Jeon
- Laboratory of Biochemistry and Cellular Engineering, Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea.
| |
Collapse
|
8
|
Schreiber A, Stühn LG, Huber MC, Geissinger SE, Rao A, Schiller SM. Self-Assembly Toolbox of Tailored Supramolecular Architectures Based on an Amphiphilic Protein Library. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900163. [PMID: 31173449 DOI: 10.1002/smll.201900163] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/15/2019] [Indexed: 06/09/2023]
Abstract
The molecular structuring of complex architectures and the enclosure of space are essential requirements for technical and living systems. Self-assembly of supramolecular structures with desired shape, size, and stability remains challenging since it requires precise regulation of physicochemical and conformational properties of the components. Here a general platform for controlled self-assembly of tailored amphiphilic elastin-like proteins into desired supramolecular protein assemblies ranging from spherical coacervates over molecularly defined twisted fibers to stable unilamellar vesicles is introduced. The described assembly protocols efficiently yield protein membrane-based compartments (PMBC) with adjustable size, stability, and net surface charge. PMBCs demonstrate membrane fusion and phase separation behavior and are able to encapsulate structurally and chemically diverse cargo molecules ranging from small molecules to naturally folded proteins. The ability to engineer tailored supramolecular architectures with defined fusion behavior, tunable properties, and encapsulated cargo paves the road for novel drug delivery systems, the design of artificial cells, and confined catalytic nanofactories.
Collapse
Affiliation(s)
- Andreas Schreiber
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79085, Freiburg, Germany
| | - Lara G Stühn
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79085, Freiburg, Germany
| | - Matthias C Huber
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79085, Freiburg, Germany
| | - Süreyya E Geissinger
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79085, Freiburg, Germany
| | - Ashit Rao
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
| | - Stefan M Schiller
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79085, Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
- IMTEK Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 103, 79110, Freiburg, Germany
- University of Freiburg, Georges-Köhler-Allee 105, 79110, Freiburg, Germany
| |
Collapse
|
9
|
Meco E, Lampe KJ. Impact of Elastin-like Protein Temperature Transition on PEG-ELP Hybrid Hydrogel Properties. Biomacromolecules 2019; 20:1914-1925. [DOI: 10.1021/acs.biomac.9b00113] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Edi Meco
- Department of Chemical Engineering, Chemical Eng., Office 117, University of Virginia, 102 Engineer’s Way, Charlottesville, Virginia 22904, United States
| | - Kyle J. Lampe
- Department of Chemical Engineering, Chemical Eng., Office 117, University of Virginia, 102 Engineer’s Way, Charlottesville, Virginia 22904, United States
| |
Collapse
|
10
|
Gonzalez-Valdivieso J, Girotti A, Muñoz R, Rodriguez-Cabello JC, Arias FJ. Self-Assembling ELR-Based Nanoparticles as Smart Drug-Delivery Systems Modulating Cellular Growth via Akt. Biomacromolecules 2019; 20:1996-2007. [DOI: 10.1021/acs.biomac.9b00206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Juan Gonzalez-Valdivieso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Raquel Muñoz
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - J. Carlos Rodriguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - F. Javier Arias
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| |
Collapse
|
11
|
Quintanilla-Sierra L, García-Arévalo C, Rodriguez-Cabello J. Self-assembly in elastin-like recombinamers: a mechanism to mimic natural complexity. Mater Today Bio 2019; 2:100007. [PMID: 32159144 PMCID: PMC7061623 DOI: 10.1016/j.mtbio.2019.100007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
The topic of self-assembled structures based on elastin-like recombinamers (ELRs, i.e., elastin-like polymers recombinantly bio-produced) has released a noticeable amount of references in the last few years. Most of them are intended for biomedical applications. In this review, a complete revision of the bibliography is carried out. Initially, the self-assembly (SA) concept is considered from a general point of view, and then ELRs are described and characterized based on their intrinsic disorder. A classification of the different self-assembled ELR-based structures is proposed based on their morphologies, paying special attention to their tentative modeling. The impact of the mechanism of SA on these biomaterials is analyzed. Finally, the implications of ELR SA in biological systems are considered.
Collapse
Affiliation(s)
| | | | - J.C. Rodriguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, 47011, Valladolid, Spain
| |
Collapse
|
12
|
Cipriani F, Krüger M, de Torre IG, Sierra LQ, Rodrigo MA, Kock L, Rodriguez-Cabello JC. Cartilage Regeneration in Preannealed Silk Elastin-Like Co-Recombinamers Injectable Hydrogel Embedded with Mature Chondrocytes in an Ex Vivo Culture Platform. Biomacromolecules 2018; 19:4333-4347. [PMID: 30346149 DOI: 10.1021/acs.biomac.8b01211] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tissue engineering for cartilage repair requires biomaterials that show rapid gelation and adequate mechanical properties. Although the use of hydrogel is the most promising biomaterial, it often lacks in rigidity and anchorage of cells when they are surrounded by synovial fluid while they are subjected to heavy loads. We developed and produced the Silk Elastin-Like co-Recombinamer (SELR), which contains both the physical interaction from elastin motifs and from silk motifs. In the first part of this work, we set up and optimized a preannealing treatment based on the evolution of silk motifs into β-sheet structures in order to fulfill the required mechanical properties of hydrogels for cartilage repair. The new preannealed SELRs (pA(EIS)2-(I5R)6) were characterized with the combination of several experimental techniques (CD, TEM, SEM, and rheology) to provide a deep insight into the material features. Finally, the regeneration properties of the pA(EIS)2-(I5R)6 hydrogel embedded with chondrocytes were evaluated. After 4 weeks of culturing in a standardized and representative ex vivo model, the biochemical and histological analysis revealed the production of glycosaminglycans and collagen. Moreover, the immunohistochemistry showed the absence of fibro-cartilage and the presence of hyaline cartilage. Hence, we conclude that the pA(EIS)2-(I5R)6 hydrogel presents improved mechanical properties while conserving the injectability, which leads to successful regeneration of hyaline cartilage in an ex vivo model.
Collapse
Affiliation(s)
- Filippo Cipriani
- Technical Proteins Nanobiotechnology S.L. , Paseo Belén 9A , 47001 Valladolid , Spain
| | - Melanie Krüger
- LifeTec Group B.V. , 5611 ZS Eindhoven , The Netherlands
| | - Israel Gonzalez de Torre
- Technical Proteins Nanobiotechnology S.L. , Paseo Belén 9A , 47001 Valladolid , Spain.,Bioforge , University of Valladolid CIBER-BNN , Paseo de Belén 19 , 47001 Valladolid , Spain
| | - Luis Quintanilla Sierra
- Bioforge , University of Valladolid CIBER-BNN , Paseo de Belén 19 , 47001 Valladolid , Spain
| | - Matilde Alonso Rodrigo
- Technical Proteins Nanobiotechnology S.L. , Paseo Belén 9A , 47001 Valladolid , Spain.,Bioforge , University of Valladolid CIBER-BNN , Paseo de Belén 19 , 47001 Valladolid , Spain
| | - Linda Kock
- LifeTec Group B.V. , 5611 ZS Eindhoven , The Netherlands
| | - José Carlos Rodriguez-Cabello
- Technical Proteins Nanobiotechnology S.L. , Paseo Belén 9A , 47001 Valladolid , Spain.,Bioforge , University of Valladolid CIBER-BNN , Paseo de Belén 19 , 47001 Valladolid , Spain
| |
Collapse
|
13
|
Fernández-Colino A, Wolf F, Rütten S, Rodríguez-Cabello JC, Jockenhoevel S, Mela P. Combining Catalyst-Free Click Chemistry with Coaxial Electrospinning to Obtain Long-Term, Water-Stable, Bioactive Elastin-Like Fibers for Tissue Engineering Applications. Macromol Biosci 2018; 18:e1800147. [PMID: 30260568 DOI: 10.1002/mabi.201800147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/09/2018] [Indexed: 11/09/2022]
Abstract
Elastic fibers are a fundamental requirement for tissue-engineered equivalents of physiologically elastic native tissues. Here, a simple one-step electrospinning approach is developed, combining i) catalyst-free click chemistry, ii) coaxial electrospinning, and iii) recombinant elastin-like polymers as a relevant class of biomaterials. Water-stable elastin-like fibers are obtained without the use of cross-linking agents, catalysts, or harmful organic solvents. The fibers can be directly exposed to an aqueous environment at physiological temperature and their morphology maintained for at least 3 months. The bioactivity of the fibers is demonstrated with human vascular cells and the potential of the process for vascular tissue engineering is shown by fabricating small-diameter tubular fibrous scaffolds. Moreover, highly porous fluffy 3D constructs are obtained without the use of specially designed collectors or sacrificial materials, further supporting their applicability in the biomedical field. Ultimately, the strategy that is developed here may be applied to other click systems, contributing to expanding their potential in medical technology.
Collapse
Affiliation(s)
- Alicia Fernández-Colino
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
| | - Frederic Wolf
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Pauwelsstrasse, 30, D-52074, Aachen, Germany
| | | | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany.,Aachen-Maastricht-Institute for Biobased Materials (AMIBM), Maastricht University, Urmonderbaan 22, 6167 RD, Geleen, The Netherlands
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074, Aachen, Germany
| |
Collapse
|
14
|
Qiu F, Chen Y, Tang C, Zhao X. Amphiphilic peptides as novel nanomaterials: design, self-assembly and application. Int J Nanomedicine 2018; 13:5003-5022. [PMID: 30214203 PMCID: PMC6128269 DOI: 10.2147/ijn.s166403] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Designer self-assembling peptides are a category of emerging nanobiomaterials which have been widely investigated in the past decades. In this field, amphiphilic peptides have received special attention for their simplicity in design and versatility in application. This review focuses on recent progress in designer amphiphilic peptides, trying to give a comprehensive overview about this special type of self-assembling peptides. By exploring published studies on several typical types of amphiphilic peptides in recent years, herein we discuss in detail the basic design, self-assembling behaviors and the mechanism of amphiphilic peptides, as well as how their nanostructures are affected by the peptide characteristics or environmental parameters. The applications of these peptides as potential nanomaterials for nanomedicine and nanotechnology are also summarized.
Collapse
Affiliation(s)
- Feng Qiu
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu 610041, China, .,Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, China, ,
| | - Yongzhu Chen
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, China, , .,Periodical Press of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengkang Tang
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, China, , .,Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, China, ,
| |
Collapse
|
15
|
Fernández-Colino A, Wolf F, Keijdener H, Rütten S, Schmitz-Rode T, Jockenhoevel S, Rodríguez-Cabello JC, Mela P. Macroporous click-elastin-like hydrogels for tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 88:140-147. [PMID: 29636129 DOI: 10.1016/j.msec.2018.03.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/15/2018] [Accepted: 03/15/2018] [Indexed: 01/13/2023]
Abstract
Elastin is a key extracellular matrix (ECM) protein that imparts functional elasticity to tissues and therefore an attractive candidate for bioengineering materials. Genetically engineered elastin-like recombinamers (ELRs) maintain inherent properties of the natural elastin (e.g. elastic behavior, bioactivity, low thrombogenicity, inverse temperature transition) while featuring precisely controlled composition, the possibility for biofunctionalization and non-animal origin. Recently the chemical modification of ELRs to enable their crosslinking via a catalyst-free click chemistry reaction, has further widened their applicability for tissue engineering. Despite these outstanding properties, the generation of macroporous click-ELR scaffolds with controlled, interconnected porosity has remained elusive so far. This significantly limits the potential of these materials as the porosity has a crucial role on cell infiltration, proliferation and ECM formation. In this study we propose a strategy to overcome this issue by adapting the salt leaching/gas foaming technique to click-ELRs. As result, macroporous hydrogels with tuned pore size and mechanical properties in the range of many native tissues were reproducibly obtained as demonstrated by rheological measurements and quantitative analysis of fluorescence, scanning electron and two-photon microscopy images. Additionally, the appropriate size and interconnectivity of the pores enabled smooth muscle cells to migrate into the click-ELR scaffolds and deposit extracellular matrix. The macroporous structure together with the elastic performance and bioactive character of ELRs, the specificity and non-toxic character of the catalyst-free click-chemistry reaction, make these scaffolds promising candidates for applications in tissue regeneration. This work expands the potential use of ELRs and click chemistry systems in general in different biomedical fields.
Collapse
Affiliation(s)
- Alicia Fernández-Colino
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany.
| | - Frederic Wolf
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany
| | - Hans Keijdener
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Pauwelsstrasse, 30, D-52074 Aachen, Germany
| | - Thomas Schmitz-Rode
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany; AMIBM-Aachen-Maastricht-Institute for Biobased Materials, Maastricht University, Urmonderbaan 22, 6167 RD, Geleen, The Netherlands
| | | | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex) at AME-Institute of Applied Medical Engineering, Helmholtz Institute-CBMS, RWTH Aachen University, Forckenbeckstr. 55, 52074 Aachen, Germany.
| |
Collapse
|
16
|
Sahoo JK, VandenBerg MA, Webber MJ. Injectable network biomaterials via molecular or colloidal self-assembly. Adv Drug Deliv Rev 2018; 127:185-207. [PMID: 29128515 DOI: 10.1016/j.addr.2017.11.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 09/16/2017] [Accepted: 11/06/2017] [Indexed: 11/19/2022]
Abstract
Self-assembly is a powerful tool to create functional materials. A specific application for which self-assembled materials are ideally suited is in creating injectable biomaterials. Contrasting with traditional biomaterials that are implanted through surgical means, injecting biomaterials through the skin offers numerous advantages, expanding the scope and impact for biomaterials in medicine. In particular, self-assembled biomaterials prepared from molecular or colloidal interactions have been frequently explored. The strategies to create these materials are varied, taking advantage of engineered oligopeptides, proteins, and nanoparticles as well as affinity-mediated crosslinking of synthetic precursors. Self-assembled materials typically facilitate injectability through two different mechanisms: i) in situ self-assembly, whereby materials would be administered in a monomeric or oligomeric form and self-assemble in response to some physiologic stimulus, or ii) self-assembled materials that, by virtue of their dynamic, non-covalent interactions, shear-thin to facilitate flow within a syringe and subsequently self-heal into its reassembled material form at the injection site. Indeed, many classes of materials are capable of being injected using a combination of these two mechanisms. Particular utility has been noted for self-assembled biomaterials in the context of tissue engineering, regenerative medicine, drug delivery, and immunoengineering. Given the controlled and multifunctional nature of many self-assembled materials demonstrated to date, we project a future where injectable self-assembled biomaterials afford improved practice in advancing healthcare.
Collapse
Affiliation(s)
- Jugal Kishore Sahoo
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA
| | - Michael A VandenBerg
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Advanced Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, IN 46556, USA; Warren Family Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, IN 46556, USA; Center for Nanoscience and Technology (NDnano), University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
17
|
Duan K, Ghosh G, Lo JF. Optimizing Multiplexed Detections of Diabetes Antibodies via Quantitative Microfluidic Droplet Array. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:10.1002/smll.201702323. [PMID: 28990274 PMCID: PMC5755373 DOI: 10.1002/smll.201702323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 08/11/2017] [Indexed: 05/02/2023]
Abstract
Sensitive, single volume detections of multiple diabetes antibodies can provide immunoprofiling and early screening of at-risk patients. To advance the state-of-the-art suspension assays for diabetes antibodies, porous hydrogel droplets are leveraged in microfluidic serpentine arrays to enhance reagent transport. This spatially multiplexed assay is applied to the detection of antibodies against insulin, glutamic acid decarboxylase, and insulinoma-associated protein 2. Optimization of assay protocol results in a shortened assay time of 2 h, with better than 20 pg mL Supporting Information detection limits across all three antibodies. Specificity and cross-reactivity tests show negligible background, nonspecific antibody-antigen, and nonspecific antibody-antibody bindings. Multiplexed detections are able to measure within 15% of target concentrations from low to high ranges. The technique enables quantifications of as little as 8000 molecules in each 500 µm droplet in a single volume, multiplexed assay format, a breakthrough necessary for the adoption of diabetes panels for clinical screening and monitoring in the future.
Collapse
Affiliation(s)
- Kai Duan
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan at Dearborn, Dearborn, MI, 48128, USA
| | - Gargi Ghosh
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan at Dearborn, Dearborn, MI, 48128, USA
| | - Joe Fujiou Lo
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan at Dearborn, Dearborn, MI, 48128, USA
| |
Collapse
|
18
|
Coletta DJ, Ibáñez-Fonseca A, Missana LR, Jammal MV, Vitelli EJ, Aimone M, Zabalza F, Issa JPM, Alonso M, Rodríguez-Cabello JC, Feldman S. Bone Regeneration Mediated by a Bioactive and Biodegradable Extracellular Matrix-Like Hydrogel Based on Elastin-Like Recombinamers. Tissue Eng Part A 2017; 23:1361-1371. [DOI: 10.1089/ten.tea.2017.0047] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Dante J. Coletta
- LABOATEM, Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | | | - Liliana R. Missana
- Experimental Pathology and Tissue Engineering Laboratory, Dental School, National Tucumán University, Tucumán, Argentina
- Tissues Laboratory, Proimi-Biotechnology-Conicet, Tucumán, Argentina
| | - María V. Jammal
- Experimental Pathology and Tissue Engineering Laboratory, Dental School, National Tucumán University, Tucumán, Argentina
- Tissues Laboratory, Proimi-Biotechnology-Conicet, Tucumán, Argentina
| | - Ezequiel J. Vitelli
- LABOATEM, Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | - Mariangeles Aimone
- LABOATEM, Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | - Facundo Zabalza
- LABOATEM, Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | | | - Matilde Alonso
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
| | | | - Sara Feldman
- LABOATEM, Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| |
Collapse
|
19
|
Changi K, Bosnjak B, Gonzalez-Obeso C, Kluger R, Rodríguez-Cabello JC, Hoffmann O, Epstein MM. Biocompatibility and immunogenicity of elastin-like recombinamer biomaterials in mouse models. J Biomed Mater Res A 2017; 106:924-934. [PMID: 29105979 DOI: 10.1002/jbm.a.36290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 10/20/2017] [Accepted: 11/02/2017] [Indexed: 12/20/2022]
Abstract
Novel thermo-sensitive elastin-like recombinamers (ELRs) containing bioactive molecules were created for use as a biomimetic biomaterial for tissue regeneration. For effective use for in vivo applications, it is essential to ensure that they do not induce adverse inflammatory, immune, or allergic responses that inhibit tissue repair. Therefore, we sought to establish a pre-clinical approach to evaluate biocompatibility in experimental mice using ELRs as a prototype biomaterial. First, we measured in vitro proliferation and cytokine production from BALB/c and C57BL/6 mouse splenocytes incubated with ELRs. Second, we used a rapid, high throughput in vivo approach in which inflammatory cells and cytokines were measured following an intraperitoneal implantation. Lastly, a subchronic in vivo approach was used in which ELRs or positive controls were subcutaneously implanted and the implantation sites were assessed for inflammation and gene expression. We found that ELRs induced mild inflammation and minimal fibrosis compared to the intense response to Vitoss. Additionally, implantation increased antigen-specific antibody titers for both groups and gene expression profiling of the implantation sites revealed the upregulation of inflammation, fibrosis, and wound healing-related genes in ELR and positive control-implanted mice compared to sham controls. These data demonstrate that ELRs appear safe for use in tissue engineering. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 924-934, 2018.
Collapse
Affiliation(s)
- K Changi
- Department of Dermatology, Laboratory of Experimental Allergy, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - B Bosnjak
- Department of Dermatology, Laboratory of Experimental Allergy, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - C Gonzalez-Obeso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, Valladolid, Spain
| | - R Kluger
- Danube Hospital Vienna, Vienna, Austria
| | - J C Rodríguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, Valladolid, Spain
| | - O Hoffmann
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - M M Epstein
- Department of Dermatology, Laboratory of Experimental Allergy, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Hydrogel-based suspension array for biomarker detection using horseradish peroxidase-mediated silver precipitation. Anal Chim Acta 2017; 999:132-138. [PMID: 29254564 DOI: 10.1016/j.aca.2017.10.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/18/2017] [Accepted: 10/26/2017] [Indexed: 01/25/2023]
Abstract
Advances in medical diagnostics and personalized therapy require robust, sensitive yet cost-effective diagnostic tools for rapid measurement of biomolecules including proteins in body fluids. State-of-the-art technologies are complex and rely on expensive or custom made detection system, and therefore, cannot be readily adapted for point-of-care (POC) analysis. The development of a novel detection platform, which leverages horseradish peroxidase (HRP)-mediated silver precipitation within antibody immobilized porosity tuned poly (ethylene) glycol diacrylate (PEGDA) hydrogel microparticles with the operational advantages of suspension arrays for sensitive quantification of biomarkers, is described. In this study, vascular endothelial growth factor (VEGF) has been used as a model protein. The silver deposition corresponded to the concentration of VEGF in solution. The detection limit of 5.2 ± 1.0 pg/mL and assay time of 2 h highlights that this assay exceeds the conventional technologies in terms of sensitivity and speed. The practical applicability of the hydrogel microparticle based detection system has been established by demonstrating the ability of the system to quantify the production of VEGF by highly aggressive (MDA-MB-231) and non-aggressive (MCF-7) breast cancer cells. The reliance on simple instrument for quantification of clinically relevant markers bolsters the adaptability of the detection platform/method in POC settings.
Collapse
|
21
|
Shieh HF, Graham CD, Brazzo JA, Zurakowski D, Fauza DO. Comparisons of human amniotic mesenchymal stem cell viability in FDA-approved collagen-based scaffolds: Implications for engineered diaphragmatic replacement. J Pediatr Surg 2017; 52:1010-1013. [PMID: 28366561 DOI: 10.1016/j.jpedsurg.2017.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/09/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND/PURPOSE We sought to examine amniotic fluid mesenchymal stem cell (afMSC) viability within two FDA-approved collagen-based scaffolds, as a prerequisite to clinical translation of afMSC-based engineered diaphragmatic repair. METHODS Human afMSCs were seeded in a human-derived collagen hydrogel and in a bovine-derived collagen sheet at 3 matching densities. Cell viability was analyzed at 1, 3, and 5days using an ATP-based 3D bioluminescence assay. Statistical comparisons were by ANOVA (P<0.05). RESULTS There was a highly significant 3-way interaction between scaffold type, seeding density, and time in 3D culture as determinants of cell viability, clearly favoring the human hydrogel (P<0.001). In both scaffolds, cell viability was highest at the highest seeding density of 150,000 cells/mL. Time in 3D culture impacted cell viability at the optimal seeding density in the human hydrogel, with the highest levels on days 1 (P<0.001) and 5 (P=0.05) with no significant effect in the bovine sheet (P=0.39-0.96). CONCLUSIONS Among clinically-approved cell delivery vehicles, mesenchymal stem cell viability is significantly enhanced in a collagen hydrogel when compared with a collagen sheet. Cell viability can be further optimized by seeding density and time in 3D culture. These data further support the regulatory viability of clinical trials of engineered diaphragmatic repair. LEVEL OF EVIDENCE N/A (animal and laboratory study).
Collapse
Affiliation(s)
- Hester F Shieh
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Christopher D Graham
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Joseph A Brazzo
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
22
|
Vila M, García A, Girotti A, Alonso M, Rodríguez-Cabello JC, González-Vázquez A, Planell JA, Engel E, Buján J, García-Honduvilla N, Vallet-Regí M. 3D silicon doped hydroxyapatite scaffolds decorated with Elastin-like Recombinamers for bone regenerative medicine. Acta Biomater 2016; 45:349-356. [PMID: 27639311 DOI: 10.1016/j.actbio.2016.09.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/20/2016] [Accepted: 09/14/2016] [Indexed: 12/19/2022]
Abstract
The current study reports on the manufacturing by rapid prototyping technique of three-dimensional (3D) scaffolds based on silicon substituted hydroxyapatite with Elastin-like Recombinamers (ELRs) functionalized surfaces. Silicon doped hydroxyapatite (Si-HA), with Ca10(PO4)5.7(SiO4)0.3(OH)1.7h0.3 nominal formula, was surface functionalized with two different types of polymers designed by genetic engineering: ELR-RGD that contain cell attachment specific sequences and ELR-SNA15/RGD with both hydroxyapatite and cells domains that interact with the inorganic phase and with the cells, respectively. These hybrid materials were subjected to in vitro assays in order to clarify if the ELRs coating improved the well-known biocompatible and bone regeneration properties of calcium phosphates materials. The in vitro tests showed that there was a total and homogeneous colonization of the 3D scaffolds by Bone marrow Mesenchymal Stromal Cells (BMSCs). In addition, the BMSCs were viable and able to proliferate and differentiate into osteoblasts. STATEMENT OF SIGNIFICANCE Bone tissue engineering is an area of increasing interest because its main applications are directly related to the rising life expectancy of the population, which promotes higher rates of several bone pathologies, so innovative strategies are needed for bone tissue regeneration therapies. Here we use the rapid prototyping technology to allow moulding ceramic 3D scaffolds and we use different bio-polymers for the functionalization of their surfaces in order to enhance the biological response. Combining the ceramic material (silicon doped hydroxyapatite, Si-HA) and the Elastin like Recombinamers (ELRs) polymers with the presence of the integrin-mediate adhesion domain alone or in combination with SNA15 peptide that possess high affinity for hydroxyapatite, provided an improved Bone marrow Mesenchymal Stromal Cells (BMSCs) differentiation into osteoblastic linkage.
Collapse
|
23
|
Miranda-Nieves D, Chaikof EL. Collagen and Elastin Biomaterials for the Fabrication of Engineered Living Tissues. ACS Biomater Sci Eng 2016; 3:694-711. [PMID: 33440491 DOI: 10.1021/acsbiomaterials.6b00250] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Collagen and elastin represent the two most predominant proteins in the body and are responsible for modulating important biological and mechanical properties. Thus, the focus of this review is the use of collagen and elastin as biomaterials for the fabrication of living tissues. Considering the importance of both biomaterials, we first propose the notion that many tissues in the human body represent a reinforced composite of collagen and elastin. In the rest of the review, collagen and elastin biosynthesis and biophysics, as well as molecular sources and biomaterial fabrication methodologies, including casting, fiber spinning, and bioprinting, are discussed. Finally, we summarize the current attempts to fabricate a subset of living tissues and, based on biochemical and biomechanical considerations, suggest that future tissue-engineering efforts consider direct incorporation of collagen and elastin biomaterials.
Collapse
Affiliation(s)
- David Miranda-Nieves
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, United States
| | - Elliot L Chaikof
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, United States.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
24
|
Piña MJ, Girotti A, Santos M, Rodríguez-Cabello JC, Arias FJ. Biocompatible ELR-Based Polyplexes Coated with MUC1 Specific Aptamers and Targeted for Breast Cancer Gene Therapy. Mol Pharm 2016; 13:795-808. [PMID: 26815223 DOI: 10.1021/acs.molpharmaceut.5b00712] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The search for new and biocompatible materials with high potential for improvement is a challenge in gene delivery applications. A cell type specific vector made of elastin-like recombinamer (ELR) and aptamers has been specifically designed for the intracellular delivery of therapeutic material for breast cancer therapy. A lysine-enriched ELR was constructed and complexed with plasmid DNA to give positively charged and stable polyplexes. Physical characterization of these polyplexes showed a particle size of around 140 nm and a zeta potential of approximately +40 mV. The incorporation of MUC1-specific aptamers into the polyplexes resulted in a slight decrease in zeta potential but increased cell transfection specificity for MCF-7 breast cancer cells with respect to a MUC1-negative tumor line. After showing the transfection ability of this aptamer-ELR vector which is facilitated mainly by macropinocytosis uptake, we demonstrated its application for suicide gene therapy using a plasmid containing the gene of the toxin PAP-S. The strategy developed in this work about using ELR as polymeric vector and aptamers as supplier of specificity to deliver therapeutic material into MUC1-positive breast cancer cells shows promising potential and continues paving the way for ELRs in the biomedical field.
Collapse
Affiliation(s)
- Maria J Piña
- Bioforge Research Group, CIBER-BBN, University of Valladolid , LUCIA, Paseo de Belén 19, 47011 Valladolid, Spain
| | - Alessandra Girotti
- Bioforge Research Group, CIBER-BBN, University of Valladolid , LUCIA, Paseo de Belén 19, 47011 Valladolid, Spain
| | - Mercedes Santos
- Bioforge Research Group, CIBER-BBN, University of Valladolid , LUCIA, Paseo de Belén 19, 47011 Valladolid, Spain
| | - J Carlos Rodríguez-Cabello
- Bioforge Research Group, CIBER-BBN, University of Valladolid , LUCIA, Paseo de Belén 19, 47011 Valladolid, Spain
| | - F Javier Arias
- Bioforge Research Group, CIBER-BBN, University of Valladolid , LUCIA, Paseo de Belén 19, 47011 Valladolid, Spain
| |
Collapse
|
25
|
Rodríguez-Cabello JC, Arias FJ, Rodrigo MA, Girotti A. Elastin-like polypeptides in drug delivery. Adv Drug Deliv Rev 2016; 97:85-100. [PMID: 26705126 DOI: 10.1016/j.addr.2015.12.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 12/12/2022]
Abstract
The use of recombinant elastin-like materials, or elastin-like recombinamers (ELRs), in drug-delivery applications is reviewed in this work. Although ELRs were initially used in similar ways to other, more conventional kinds of polymeric carriers, their unique properties soon gave rise to systems of unparalleled functionality and efficiency, with the stimuli responsiveness of ELRs and their ability to self-assemble readily allowing the creation of advanced systems. However, their recombinant nature is likely the most important factor that has driven the current breakthrough properties of ELR-based delivery systems. Recombinant technology allows an unprecedented degree of complexity in macromolecular design and synthesis. In addition, recombinant materials easily incorporate any functional domain present in natural proteins. Therefore, ELR-based delivery systems can exhibit complex interactions with both their drug load and the tissues and cells towards which this load is directed. Selected examples, ranging from highly functional nanocarriers to macrodepots, will be presented.
Collapse
|
26
|
Girotti A, Orbanic D, Ibáñez-Fonseca A, Gonzalez-Obeso C, Rodríguez-Cabello JC. Recombinant Technology in the Development of Materials and Systems for Soft-Tissue Repair. Adv Healthc Mater 2015; 4:2423-55. [PMID: 26172311 DOI: 10.1002/adhm.201500152] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/04/2015] [Indexed: 12/16/2022]
Abstract
The field of biomedicine is constantly investing significant research efforts in order to gain a more in-depth understanding of the mechanisms that govern the function of body compartments and to develop creative solutions for the repair and regeneration of damaged tissues. The main overall goal is to develop relatively simple systems that are able to mimic naturally occurring constructs and can therefore be used in regenerative medicine. Recombinant technology, which is widely used to obtain new tailored synthetic genes that express polymeric protein-based structures, now offers a broad range of advantages for that purpose by permitting the tuning of biological and mechanical properties depending on the intended application while simultaneously ensuring adequate biocompatibility and biodegradability of the scaffold formed by the polymers. This Progress Report is focused on recombinant protein-based materials that resemble naturally occurring proteins of interest for use in soft tissue repair. An overview of recombinant biomaterials derived from elastin, silk, collagen and resilin is given, along with a description of their characteristics and suggested applications. Current endeavors in this field are continuously providing more-improved materials in comparison with conventional ones. As such, a great effort is being made to put these materials through clinical trials in order to favor their future use.
Collapse
Affiliation(s)
- Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology); CIBER-BBN; University of Valladolid, Edificio LUCIA; Paseo de Belén, 19 47011 Valladolid Spain
| | - Doriana Orbanic
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology); CIBER-BBN; University of Valladolid, Edificio LUCIA; Paseo de Belén, 19 47011 Valladolid Spain
| | - Arturo Ibáñez-Fonseca
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology); CIBER-BBN; University of Valladolid, Edificio LUCIA; Paseo de Belén, 19 47011 Valladolid Spain
| | - Constancio Gonzalez-Obeso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology); CIBER-BBN; University of Valladolid, Edificio LUCIA; Paseo de Belén, 19 47011 Valladolid Spain
| | - José Carlos Rodríguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology); CIBER-BBN; University of Valladolid, Edificio LUCIA; Paseo de Belén, 19 47011 Valladolid Spain
| |
Collapse
|
27
|
Piña MJ, Alex SM, Arias FJ, Santos M, Rodriguez-Cabello JC, Ramesan RM, Sharma CP. Elastin-like recombinamers with acquired functionalities for gene-delivery applications. J Biomed Mater Res A 2015; 103:3166-78. [DOI: 10.1002/jbm.a.35455] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/26/2015] [Accepted: 03/10/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Maria J. Piña
- Bioforge Research Group, University of Valladolid, CIBER-BBN; Valladolid 47011 Spain
| | - Susan M. Alex
- Biosurface Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura; Thiruvananthapuram Kerala 695 012 India
| | - Francisco J. Arias
- Bioforge Research Group, University of Valladolid, CIBER-BBN; Valladolid 47011 Spain
| | - Mercedes Santos
- Bioforge Research Group, University of Valladolid, CIBER-BBN; Valladolid 47011 Spain
| | | | - Rekha M. Ramesan
- Biosurface Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura; Thiruvananthapuram Kerala 695 012 India
| | - Chandra P. Sharma
- Biosurface Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Poojappura; Thiruvananthapuram Kerala 695 012 India
| |
Collapse
|
28
|
Glassman MJ, Olsen BD. End Block Design Modulates the Assembly and Mechanics of Thermoresponsive, Dual-Associative Protein Hydrogels. Macromolecules 2015. [DOI: 10.1021/ma502494s] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Matthew J. Glassman
- Department
of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts
Ave, Room 66-153, Cambridge, Massachusetts 02139, United States
| | - Bradley D. Olsen
- Department
of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts
Ave, Room 66-153, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
29
|
Abstract
Engineered diaphragmatic repair is emblematic of perinatal regenerative medicine and of the fetal tissue engineering concept. The alternative of a cellularized graft for the repair of a congenital diaphragmatic defect in the neonatal period is both biologically justifiable by the mechanisms behind diaphragmatic hernia recurrence as well as an ideal match for fetal mesenchymal stem cell-based constructs. It has been among the most developed experimental pursuits in neonatal tissue engineering, of which clinical application should be forthcoming.
Collapse
Affiliation(s)
- Dario O Fauza
- Department of Surgery, Boston Children׳s Hospital, 300 Longwood Ave, Fegan 3, Boston, Massachusetts 02115; Department of Surgery, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
30
|
Wang E, Desai MS, Heo K, Lee SW. Graphene-based materials functionalized with elastin-like polypeptides. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:2223-2229. [PMID: 24512378 DOI: 10.1021/la404333b] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Graphene-based materials commonly require functionalization for biological applications in order to control their physical/colloidal properties and to introduce additional capabilities, such as stimuli-responsiveness and affinity to specific biomolecules. Here, we functionalized CVD-grown graphene and graphene oxide with a genetically engineered elastin-like polypeptide fused to a graphene binding peptide and then showed that the resulting hybrid materials exhibit thermo- and photoresponsive behaviors. Furthermore, we demonstrate that our genetic engineering strategy allows for the facile introduction of bioactivity to reduced graphene oxide. The stimuli-responsiveness and genetic tunability of our graphene-protein nanocomposites are attractive for addressing future biomedical applications.
Collapse
Affiliation(s)
- Eddie Wang
- Department of Bioengineering, University of California, Berkeley , Berkeley, California 94720, United States
| | | | | | | |
Collapse
|
31
|
Kumar VA, Martinez AW, Caves JM, Naik N, Haller CA, Chaikof EL. Microablation of collagen-based substrates for soft tissue engineering. Biomed Mater 2014; 9:011002. [PMID: 24457193 DOI: 10.1088/1748-6041/9/1/011002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Noting the abundance and importance of collagen as a biomaterial, we have developed a facile method for the production of a dense fibrillar extracellular matrix mimicking collagen-elastin hybrids with tunable mechanical properties. Through the use of excimer-laser technology, we have optimized conditions for the ablation of collagen lamellae without denaturation of protein, maintenance of fibrillar ultrastructure and preservation of native D-periodicity. Strengths of collagen-elastin hybrids ranged from 0.6 to 13 MPa, elongation at break from 9 to 70% and stiffness from 2.9 to 94 MPa, allowing for the design of a wide variety of tissue specific scaffolds. Further, large (centimeter scale) lamellae can be fabricated and embedded with recombinant elastin to generate collagen-elastin hybrids. Exposed collagen in hybrids act as cell adhesive sites for rat mesenchymal stem cells that conform to ablate waveforms. The ability to modulate these features allows for the generation of a class of biopolymers that can architecturally and physiologically replicate native tissue.
Collapse
Affiliation(s)
- Vivek A Kumar
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, MA 02215, USA. Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332, USA
| | | | | | | | | | | |
Collapse
|
32
|
Shang Y, Yan Y, Hou X. Stimuli responsive elastin-like polypeptides and applications in medicine and biotechnology. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2013; 25:101-20. [DOI: 10.1080/09205063.2013.841073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
33
|
Kumar VA, Caves JM, Haller CA, Dai E, Li L, Grainger S, Chaikof EL. Acellular vascular grafts generated from collagen and elastin analogs. Acta Biomater 2013; 9:8067-74. [PMID: 23743129 PMCID: PMC3733560 DOI: 10.1016/j.actbio.2013.05.024] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/14/2013] [Accepted: 05/22/2013] [Indexed: 01/10/2023]
Abstract
Tissue-engineered vascular grafts require long fabrication times, in part due to the requirement of cells from a variety of cell sources to produce a robust, load-bearing extracellular matrix. Herein, we propose a design strategy for the fabrication of tubular conduits comprising collagen fiber networks and elastin-like protein polymers to mimic native tissue structure and function. Dense fibrillar collagen networks exhibited an ultimate tensile strength (UTS) of 0.71±0.06 MPa, strain to failure of 37.1±2.2% and Young's modulus of 2.09±0.42 MPa, comparing favorably to a UTS and a Young's modulus for native blood vessels of 1.4-11.1 MPa and 1.5±0.3 MPa, respectively. Resilience, a measure of recovered energy during unloading of matrices, demonstrated that 58.9±4.4% of the energy was recovered during loading-unloading cycles. Rapid fabrication of multilayer tubular conduits with maintenance of native collagen ultrastructure was achieved with internal diameters ranging between 1 and 4mm. Compliance and burst pressures exceeded 2.7±0.3%/100 mmHg and 830±131 mmHg, respectively, with a significant reduction in observed platelet adherence as compared to expanded polytetrafluoroethylene (ePTFE; 6.8±0.05×10(5) vs. 62±0.05×10(5) platelets mm(-2), p<0.01). Using a rat aortic interposition model, early in vivo responses were evaluated at 2 weeks via Doppler ultrasound and CT angiography with immunohistochemistry confirming a limited early inflammatory response (n=8). Engineered collagen-elastin composites represent a promising strategy for fabricating synthetic tissues with defined extracellular matrix content, composition and architecture.
Collapse
Affiliation(s)
- Vivek A. Kumar
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, MA 02215
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332
| | - Jeffrey M. Caves
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, MA 02215
| | - Carolyn A. Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, MA 02215
| | - Erbin Dai
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Liying Li
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Stephanie Grainger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, MA 02215
| | - Elliot L. Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
- Wyss Institute of Biologically Inspired Engineering of Harvard University, Boston, MA 02215
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332
| |
Collapse
|
34
|
Sago Starch-Mixed Low-Density Polyethylene Biodegradable Polymer: Synthesis and Characterization. ACTA ACUST UNITED AC 2013. [DOI: 10.1155/2013/365380] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This research focuses on synthesis and characterization of sago starch-mixed LDPE biodegradable polymer. Firstly, the effect of variation of starch content on mechanical property (elongation at break and Young’s modulus) and biodegradability of the polymer was studied. The LDPE was combined with 10%, 30%, 50%, and 70% of sago for this study. Then how the cross-linking with trimethylolpropane triacrylate (TMPTA) and electron beam (EB) irradiation influence the mechanical and thermal properties of the polymer was investigated. In the 2nd study, to avoid overwhelming of data LDPE polymer was incorporated with only 50% of starch. The starch content had direct influence on mechanical property and biodegradability of the polymer. The elongation at break decreased with increase of starch content, while Young’s modulus and mass loss (i.e., degradation) were found to increase with increase of starch content. Increase of cross-linker (TMPTA) and EB doses also resulted in increased Young’s modulus of the polymer. However, both cross-linking and EB irradiation processes rendered lowering of polymer’s melting temperature. In conclusion, starch content and modification processes play significant roles in controlling mechanical, thermal, and degradation properties of the starch-mixed LDPE synthetic polymer, thus providing the opportunity to modulate the polymer properties for tailored applications.
Collapse
|
35
|
Khan FA, Rabong C, Jordis U, Phopase J. Surprising behavior of NXO-peptides toward the lithium hydroxide solvolysis. Tetrahedron Lett 2013. [DOI: 10.1016/j.tetlet.2013.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Wang E, Desai MS, Lee SW. Light-controlled graphene-elastin composite hydrogel actuators. NANO LETTERS 2013; 13:2826-30. [PMID: 23647361 PMCID: PMC3737518 DOI: 10.1021/nl401088b] [Citation(s) in RCA: 321] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Hydrogels actuators (HAs) that can reversibly respond to stimuli have applications in diverse fields. However, faster response rates and improved control over actuation timing and location are required to fulfill their potential. To address these criteria, we synthesized near-infrared light-driven HAs by interfacing genetically engineered elastin-like polypeptides with reduced-graphene oxide sheets. The resulting nanocomposites exhibited rapid and tunable motions controlled by light position, intensity, and path, including finger-like flexing and crawling. This work demonstrates the ability of rationally designed proteins to be combined with synthetic nanoparticles for the creation of macroscale functional materials.
Collapse
Affiliation(s)
- Eddie Wang
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 USA
| | - Malav S. Desai
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 USA
| | - Seung-Wuk Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, USA.Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 USA
| |
Collapse
|
37
|
Le DHT, Hanamura R, Pham DH, Kato M, Tirrell DA, Okubo T, Sugawara-Narutaki A. Self-assembly of elastin-mimetic double hydrophobic polypeptides. Biomacromolecules 2013; 14:1028-34. [PMID: 23495825 DOI: 10.1021/bm301887m] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have constructed a novel class of "double-hydrophobic" block polypeptides based on the hydrophobic domains found in native elastin, an extracellular matrix protein responsible for the elasticity and resilience of tissues. The block polypeptides comprise proline-rich poly(VPGXG) and glycine-rich poly(VGGVG), both of which dehydrate at higher temperature but form distinct secondary structures, β-turn and β-sheet respectively. In water at 45 °C, the block polypeptides initially assemble into nanoparticles rich in β-turn structures, which further connect into long (>10 μm), beaded nanofibers along with the increase in the β-sheet content. The nanofibers obtained are well-dispersed in water, and show thermoresponsive properties. Polypeptides comprising each block component assemble into different morphologies, showing that the conjugation of poly(VPGXG) and poly(VGGVG) plays a role for beaded fiber formation. These results may provide innovative ideas for designing peptide-based materials but also opportunities for developing novel materials useful for tissue engineering and drug delivery systems.
Collapse
Affiliation(s)
- Duc H T Le
- Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Tokyo 113-8656, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
García-Arévalo C, Bermejo-Martín JF, Rico L, Iglesias V, Martín L, Rodríguez-Cabello JC, Arias FJ. Immunomodulatory Nanoparticles from Elastin-Like Recombinamers: Single-Molecules for Tuberculosis Vaccine Development. Mol Pharm 2013; 10:586-97. [DOI: 10.1021/mp300325v] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Carmen García-Arévalo
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| | - Jesús F. Bermejo-Martín
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Lucia Rico
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Verónica Iglesias
- Infection and Immunity Medical Research Unit (IMI), Microbiology
Department, Hospital Clínico Universitario-IECSCYL, Ramón y Cajal 3, 47005 Valladolid, Spain
| | - Laura Martín
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| | | | - F. Javier Arias
- Bioforge Group, University of Valladolid, CIBER-BBN, Paseo de Belén
11, 47011 Valladolid, Spain
| |
Collapse
|
39
|
Ravi S, Caves JM, Martinez AW, Haller CA, Chaikof EL. Incorporation of fibronectin to enhance cytocompatibility in multilayer elastin-like protein scaffolds for tissue engineering. J Biomed Mater Res A 2012; 101:1915-25. [PMID: 23225639 DOI: 10.1002/jbm.a.34484] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/17/2012] [Accepted: 09/24/2012] [Indexed: 11/07/2022]
Abstract
Recombinant, elastin-like protein (ELP) polymers are of significant interest for the engineering of compliant, resilient soft tissues due to a wide range of tunable mechanical properties, biostability, and biocompatibility. Here, we enhance endothelial cell (EC) and mesenchymal stem cell compatibility with ELP constructs by addition of fibronectin (Fn) to the surface or bulk of ELP hydrogels. We find that cell adhesion, proliferation, and migration can be modulated by Fn addition. Adsorption of Fn to the hydrogel surface is more efficient than bulk blending. Surface immobilization of Fn by genipin crosslinking leads to stability without loss of bioactivity. Gels of varying mechanical modulus do not alter cell adhesion, proliferation, and migration in the range we investigate. However, more compliant gels promote an EC morphology suggesting tubulogenesis or network formation, whereas stiffer gels promote cobblestone morphology. Multilayer structures consisting of thin ELP sheets reinforced with collagen microfiber are fabricated and laminated through the culture of MSCs at layer interfaces. High cell viability in the resulting three-dimensional constructs suggests the applicability of Fn to the design of strong, resilient artificial blood vessels and other soft tissue replacements.
Collapse
Affiliation(s)
- Swathi Ravi
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia 30332, USA
| | | | | | | | | |
Collapse
|
40
|
Krishna UM, Martinez AW, Caves JM, Chaikof EL. Hydrazone self-crosslinking of multiphase elastin-like block copolymer networks. Acta Biomater 2012; 8:988-97. [PMID: 22154858 DOI: 10.1016/j.actbio.2011.11.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 11/12/2011] [Accepted: 11/21/2011] [Indexed: 11/29/2022]
Abstract
Biosynthetic strategies for the production of recombinant elastin-like protein (ELP) triblock copolymers have resulted in elastomeric protein hydrogels, formed through rapid physical crosslinking upon warming of concentrated solutions. However, the strength of physically crosslinked networks can be limited, and options for non-toxic chemical crosslinking of these networks are not optimal. In this report, we modify two recombinant elastin-like proteins with aldehyde and hydrazide functionalities. When combined, these modified recombinant proteins self-crosslink through hydrazone bonding without requiring initiators or producing by-products. Crosslinked materials are evaluated for water content and swelling upon hydration, and subject to tensile and compressive mechanical tests. Hydrazone crosslinking is a viable method for increasing the mechanical strength of elastin-like protein polymers, in a manner that is likely to lend itself to the biocompatible in situ formation of chemically and physically crosslinked ELP hydrogels.
Collapse
|
41
|
Maleimide-thiol coupling of a bioactive peptide to an elastin-like protein polymer. Acta Biomater 2012; 8:627-35. [PMID: 22061108 DOI: 10.1016/j.actbio.2011.10.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/29/2011] [Accepted: 10/18/2011] [Indexed: 12/11/2022]
Abstract
Recombinant elastin-like protein (ELP) polymers display several favorable characteristics for tissue repair and replacement as well as drug delivery applications. However, these materials are derived from peptide sequences that do not lend themselves to cell adhesion, migration, or proliferation. This report describes the chemoselective ligation of peptide linkers bearing the bioactive RGD sequence to the surface of ELP hydrogels. Initially, cystamine is conjugated to ELP, followed by the temperature-driven formation of elastomeric ELP hydrogels. Cystamine reduction produces reactive thiols that are coupled to the RGD peptide linker via a terminal maleimide group. Investigations into the behavior of endothelial cells and mesenchymal stem cells on the RGD-modified ELP hydrogel surface reveal significantly enhanced attachment, spreading, migration and proliferation. Attached endothelial cells display a quiescent phenotype.
Collapse
|
42
|
|
43
|
Gomes S, Leonor IB, Mano JF, Reis RL, Kaplan DL. Natural and Genetically Engineered Proteins for Tissue Engineering. Prog Polym Sci 2012; 37:1-17. [PMID: 22058578 PMCID: PMC3207498 DOI: 10.1016/j.progpolymsci.2011.07.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
To overcome the limitations of traditionally used autografts, allografts and, to a lesser extent, synthetic materials, there is the need to develop a new generation of scaffolds with adequate mechanical and structural support, control of cell attachment, migration, proliferation and differentiation and with bio-resorbable features. This suite of properties would allow the body to heal itself at the same rate as implant degradation. Genetic engineering offers a route to this level of control of biomaterial systems. The possibility of expressing biological components in nature and to modify or bioengineer them further, offers a path towards multifunctional biomaterial systems. This includes opportunities to generate new protein sequences, new self-assembling peptides or fusions of different bioactive domains or protein motifs. New protein sequences with tunable properties can be generated that can be used as new biomaterials. In this review we address some of the most frequently used proteins for tissue engineering and biomedical applications and describe the techniques most commonly used to functionalize protein-based biomaterials by combining them with bioactive molecules to enhance biological performance. We also highlight the use of genetic engineering, for protein heterologous expression and the synthesis of new protein-based biopolymers, focusing the advantages of these functionalized biopolymers when compared with their counterparts extracted directly from nature and modified by techniques such as physical adsorption or chemical modification.
Collapse
Affiliation(s)
- Sílvia Gomes
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal
| | | | | | | | | |
Collapse
|
44
|
Girotti A, Fernández-Colino A, López IM, Rodríguez-Cabello JC, Arias FJ. Elastin-like recombinamers: Biosynthetic strategies and biotechnological applications. Biotechnol J 2011; 6:1174-86. [DOI: 10.1002/biot.201100116] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/16/2011] [Accepted: 07/28/2011] [Indexed: 01/02/2023]
|
45
|
Caves JM, Cui W, Wen J, Kumar VA, Haller CA, Chaikof EL. Elastin-like protein matrix reinforced with collagen microfibers for soft tissue repair. Biomaterials 2011; 32:5371-9. [PMID: 21550111 PMCID: PMC3207740 DOI: 10.1016/j.biomaterials.2011.04.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 04/05/2011] [Indexed: 10/18/2022]
Abstract
Artificial composites designed to mimic the structure and properties of native extracellular matrix may lead to acellular materials for soft tissue repair and replacement, which display mechanical strength, stiffness, and resilience resembling native tissue. We describe the fabrication of thin lamellae consisting of continuous collagen microfiber embedded at controlled orientations and densities in a recombinant elastin-like protein polymer matrix. Multilamellar stacking affords flexible, protein-based composite sheets whose properties are dependent upon both the elastomeric matrix and collagen content and organization. Sheets are produced with properties that range over 13-fold in elongation to break (23-314%), six-fold in Young's modulus (5.3-33.1 MPa), and more than two-fold in tensile strength (1.85-4.08 MPa), exceeding that of a number of native human tissues, including urinary bladder, pulmonary artery, and aorta. A sheet approximating the mechanical response of human abdominal wall fascia is investigated as a fascial substitute for ventral hernia repair. Protein-based composite patches prevent hernia recurrence in Wistar rats over an 8-week period with new tissue formation and sustained structural integrity.
Collapse
Affiliation(s)
- Jeffrey M. Caves
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332
| | - Wanxing Cui
- Department of Surgery, Emory University, Atlanta, GA 30322
| | - Jing Wen
- Department of Surgery, Emory University, Atlanta, GA 30322
| | - Vivek A. Kumar
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332
| | - Carolyn A. Haller
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Elliot L. Chaikof
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332
- Department of Surgery, Emory University, Atlanta, GA 30322
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02215
| |
Collapse
|
46
|
Topcic D, Kim W, Holien JK, Jia F, Armstrong PC, Hohmann JD, Straub A, Krippner G, Haller CA, Domeij H, Hagemeyer CE, Parker MW, Chaikof EL, Peter K. An activation-specific platelet inhibitor that can be turned on/off by medically used hypothermia. Arterioscler Thromb Vasc Biol 2011; 31:2015-23. [PMID: 21659646 DOI: 10.1161/atvbaha.111.226241] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Therapeutic hypothermia is successfully used, for example, in cardiac surgery to protect organs from ischemia. Cardiosurgical procedures, especially in combination with extracorporeal circulation, and hypothermia itself are potentially prothrombotic. Despite the obvious need, the long half-life of antiplatelet drugs and thus the risk of postoperative bleedings have restricted their use in cardiac surgery. We describe here the design and testing of a unique recombinant hypothermia-controlled antiplatelet fusion protein with the aim of providing increased safety of hypothermia, as well as cardiac surgery. METHODS AND RESULTS An elastin-mimetic polypeptide was fused to an activation-specific glycoprotein (GP) IIb/IIIa-blocking single-chain antibody. In silico modeling illustrated the sterical hindrance of a β-spiral conformation of elastin-mimetic polypeptide preventing the single-chain antibody from inhibiting GPIIb/IIIa at 37°C. Circular dichroism spectra demonstrated reverse temperature transition, and flow cytometry showed binding to and blocking of GPIIb/IIIa at hypothermic body temperature (≤32°C) but not at normal body temperature. In vivo thrombosis in mice was selectively inhibited at hypothermia but not at 37°C. CONCLUSIONS This is the first description of a broadly applicable pharmacological strategy by which the activity of a potential drug can be controlled by temperature. In particular, this drug steerability may provide substantial benefits for antiplatelet therapy.
Collapse
Affiliation(s)
- Denijal Topcic
- Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nair KP, Breedveld V, Weck M. Multiresponsive Reversible Polymer Networks Based on Hydrogen Bonding and Metal Coordination. Macromolecules 2011. [DOI: 10.1021/ma102462y] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Marcus Weck
- Department of Chemistry and Molecular Design Institute, New York University, New York, New York 10003, United States
| |
Collapse
|
48
|
Rodríguez-Cabello JC, Martín L, Girotti A, García-Arévalo C, Arias FJ, Alonso M. Emerging applications of multifunctional elastin-like recombinamers. Nanomedicine (Lond) 2011; 6:111-22. [PMID: 21182423 DOI: 10.2217/nnm.10.141] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Elastin-like recombinamers have grown in popularity in the field of protein-inspired biomimetic materials and have found widespread use in biomedical applications. Modern genetic-engineering techniques have allowed the design of multifunctional materials with an extraordinary control over their architecture and physicochemical properties, such as stimuli-responsiveness, monodispersity, biocompatibility or self-assembly, amongst others. Indeed, these materials are playing an increasingly important role in a diverse range of applications, such as drug delivery, tissue engineering and 'smart' systems. Herein, we review some of the most interesting examples of recent advances and progressive applications of elastin-like recombinamers in biomaterial and nano-engineering sciences in recent years.
Collapse
Affiliation(s)
- J Carlos Rodríguez-Cabello
- Bioforge Group, University of Valladolid, CIBER-BBN, Edificio I+D, Paseo de Belén 11, 47011, Valladolid, Spain.
| | | | | | | | | | | |
Collapse
|
49
|
Waterhouse A, Wise SG, Ng MKC, Weiss AS. Elastin as a nonthrombogenic biomaterial. TISSUE ENGINEERING PART B-REVIEWS 2011; 17:93-9. [PMID: 21166482 DOI: 10.1089/ten.teb.2010.0432] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Surface-induced thrombosis is a significant issue for artificial blood-contacting materials used in the treatment of cardiovascular diseases. The development of biomaterials and tissue-engineered constructs that mimic the vasculature represents a way to overcome this problem. Elastin is an extracellular matrix macromolecule that imparts arterial elasticity where it comprises up to 50% of the nonhydrated mass of the vessel. In addition to its critical role in maintaining vessel integrity and elastic properties under pulsatile flow, elastin plays an important role in signaling and regulating luminal endothelial cells and smooth muscle cells in the arterial wall. Despite its well-established significance in the vasculature and its growing use as a biomaterial in tissue engineering, the hemocompatibility of elastin is often overlooked. Past studies pointing to the potential of arterial elastin and decellularized elastin as nonthrombogenic materials have begun to be realized, with elastin scaffolds and coatings displaying increased hemocomptibility. This review explores the mechanisms of elastin's nonthrombogenicity and highlights the current problems limiting its wider application as a biomaterial. We discuss the benefits of constructing biomaterials encompassing the relevant mechanical and biological features of elastin to provide enhanced hemocompatibility to biomaterials.
Collapse
Affiliation(s)
- Anna Waterhouse
- School of Molecular Bioscience, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
50
|
Kim W, Chaikof EL. Recombinant elastin-mimetic biomaterials: Emerging applications in medicine. Adv Drug Deliv Rev 2010; 62:1468-78. [PMID: 20441783 PMCID: PMC2937194 DOI: 10.1016/j.addr.2010.04.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 04/15/2010] [Accepted: 04/17/2010] [Indexed: 12/11/2022]
Abstract
Biomaterials derived from protein-based block copolymers are increasingly investigated for potential application in medicine. In particular, recombinant elastin block copolymers provide significant opportunities to modulate material microstructure and can be processed in various forms, including particles, films, gels, and fiber networks. As a consequence, biological and mechanical responses of elastin-based biomaterials are tunable through precise control of block size and amino acid sequence. In this review, the synthesis of a set of elastin-mimetic triblock copolymers and their diverse processing methods for generating material platforms currently applied in medicine will be discussed.
Collapse
Affiliation(s)
- Wookhyun Kim
- Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, GA 30332
- Department of Surgery, Emory University, Atlanta, GA
| | - Elliot L. Chaikof
- Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, GA 30332
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30322
- Department of Surgery, Emory University, Atlanta, GA
| |
Collapse
|