501
|
Stella JA, D'Amore A, Wagner WR, Sacks MS. On the biomechanical function of scaffolds for engineering load-bearing soft tissues. Acta Biomater 2010; 6:2365-81. [PMID: 20060509 DOI: 10.1016/j.actbio.2010.01.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Revised: 12/18/2009] [Accepted: 01/04/2010] [Indexed: 11/16/2022]
Abstract
Replacement or regeneration of load-bearing soft tissues has long been the impetus for the development of bioactive materials. While maturing, current efforts continue to be confounded by our lack of understanding of the intricate multi-scale hierarchical arrangements and interactions typically found in native tissues. The current state of the art in biomaterial processing enables a degree of controllable microstructure that can be used for the development of model systems to deduce fundamental biological implications of matrix morphologies on cell function. Furthermore, the development of computational frameworks which allow for the simulation of experimentally derived observations represents a positive departure from what has mostly been an empirically driven field, enabling a deeper understanding of the highly complex biological mechanisms we wish to ultimately emulate. Ongoing research is actively pursuing new materials and processing methods to control material structure down to the micro-scale to sustain or improve cell viability, guide tissue growth, and provide mechanical integrity, all while exhibiting the capacity to degrade in a controlled manner. The purpose of this review is not to focus solely on material processing but to assess the ability of these techniques to produce mechanically sound tissue surrogates, highlight the unique structural characteristics produced in these materials, and discuss how this translates to distinct macroscopic biomechanical behaviors.
Collapse
Affiliation(s)
- John A Stella
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | |
Collapse
|
502
|
Gungormus M, Branco M, Fong H, Schneider JP, Tamerler C, Sarikaya M. Self assembled bi-functional peptide hydrogels with biomineralization-directing peptides. Biomaterials 2010; 31:7266-74. [PMID: 20591477 DOI: 10.1016/j.biomaterials.2010.06.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 06/01/2010] [Indexed: 11/30/2022]
Abstract
A peptide-based hydrogel has been designed that directs the formation of hydroxyapatite. MDG1, a twenty-seven residue peptide, undergoes triggered folding to form an unsymmetrical beta-hairpin that self-assembles in response to an increase in solution ionic strength to yield a mechanically rigid, self supporting hydrogel. The C-terminal portion of MDG1 contains a heptapeptide (MLPHHGA) capable of directing the mineralization process. Circular dichroism spectroscopy indicates that the peptide folds and assembles to form a hydrogel network rich in beta-sheet secondary structure. Oscillatory rheology indicates that the hydrogel is mechanically rigid (G' 2500Pa) before mineralization. In separate experiments, mineralization was induced both biochemically and with cementoblast cells. Mineralization-domain had little effect on the mechanical rigidity of the gel. SEM and EDXS show that MDG1 gels are capable of directing the formation of hydroxapatite. Control hydrogels, prepared by peptides either lacking the mineral-directing portion or reversing its sequence, indicated that the heptapeptide is necessary and its actions are sequence specific.
Collapse
Affiliation(s)
- Mustafa Gungormus
- Materials Science and Engineering, University of Washington, Seattle, WA, USA.
| | | | | | | | | | | |
Collapse
|
503
|
Abstract
The phase imaging atomic force microscopy is a powerful tool in surface characterization of the biomaterials, and the resulting phase image is able to detect chemical variation and reveal more detailed surface properties than the morphological image. However, the chemical- and morphological-dependent phase images were still not distinguished well. In order to better understand actual occurring phase images, we examined non-carious human maxillary incisor, microphase separated polyurethane and self-assembling peptide nanofibres. We herein reported that phase image mainly plotted the morphological change: the phase peak corresponding to the morphological valley, and the morphological peak to the phase valley, and exhibited fine surface structures of materials. The chemical-dependent phase contrast was generally masked by their inherent roughness. For the sample being very rough and having great phase separation, its chemical-dependent phase contrast could be detected at the hard tapping mode ('Amp. Ref. "set point ratio"': -0.4 to -0.8), for the sample with medium roughness only at the light tapping mode ('Amp. Ref.': -0.1 to -0.4). These results will help us understand and determine actual occurring phase images of natural or fabricated biomaterials, even, other materials.
Collapse
Affiliation(s)
- Z Ye
- Institute for NanoBiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Science Park No.1, Ke Yuan 4th St., Gao Peng Road, Hi-tech Industrial Development Zone, Chengdu, 610041, Sichuan, China
| | | |
Collapse
|
504
|
Ramachandran S, Taraban MB, Trewhella J, Gryczynski I, Gryczynski Z, Yu YB. Effect of temperature during assembly on the structure and mechanical properties of peptide-based materials. Biomacromolecules 2010; 11:1502-6. [PMID: 20481580 PMCID: PMC2893577 DOI: 10.1021/bm100138m] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mutually complementary, self-repulsive oligopeptide pairs were designed to coassemble into viscoelastic hydrogels. Peptide engineering was combined with biophysical techniques to investigate the effects of temperature on the structural and mechanical properties of the resulting hydrogels. Biophysical characterizations, including dynamic rheometry, small-angle X-ray scattering (SAXS), and fluorescence spectroscopy, were used to investigate hydrogelation at the bulk, fiber, and molecular levels, respectively. It has been found that temperature has a significant effect on the structure and mechanical properties of peptide-based biomaterials. Oligopeptide fibers assembled at 25 degrees C are formed faster and are two times thicker, and the resulting material is mechanically seven times stronger than that assembled at 5 degrees C.
Collapse
Affiliation(s)
- Sivakumar Ramachandran
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Mohali-160062, India
| | - Marc B. Taraban
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jill Trewhella
- School of Molecular and Microbial Biosciences, The University of Sydney, NSW 2006, Australia
| | - Ignacy Gryczynski
- Center for Commercialization of Fluorescence Technologies Department of Molecular Biology and Immunology, Health Science Center, University of North Texas, Fort Worth, TX 76107, USA
| | - Zygmunt Gryczynski
- Center for Commercialization of Fluorescence Technologies Department of Molecular Biology and Immunology, Health Science Center, University of North Texas, Fort Worth, TX 76107, USA
| | - Yihua Bruce Yu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
505
|
Mehta G, Williams CM, Alvarez L, Lesniewski M, Kamm RD, Griffith LG. Synergistic effects of tethered growth factors and adhesion ligands on DNA synthesis and function of primary hepatocytes cultured on soft synthetic hydrogels. Biomaterials 2010; 31:4657-71. [PMID: 20304480 PMCID: PMC2872479 DOI: 10.1016/j.biomaterials.2010.01.138] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Accepted: 01/27/2010] [Indexed: 01/01/2023]
Abstract
The composition, presentation, and spatial orientation of extracellular matrix molecules and growth factors are key regulators of cell behavior. Here, we used self-assembling peptide nanofiber gels as a modular scaffold to investigate how fibronectin-derived adhesion ligands and different modes of epidermal growth factor (EGF) presentation synergistically regulate multiple facets of primary rat hepatocyte behavior in the context of a soft gel. In the presence of soluble EGF, inclusion of dimeric RGD and the heparin binding domain from fibronectin (HB) increased hepatocyte aggregation, spreading, and metabolic function compared to unmodified gels or gels modified with a single motif, but unlike rigid substrates, gels failed to induce DNA synthesis. Tethered EGF dramatically stimulated cell aggregation and spreading under all adhesive ligand conditions and also preserved metabolic function. Surprisingly, tethered EGF elicited DNA synthesis on gels with RGD and HB. Phenotypic differences between soluble and tethered EGF stimulation of cells on peptide gels are correlated with differences in expression and phosphorylation the EGF receptor and its heterodimerization partner ErbB2, and activation of the downstream signaling node ERK1/2. These modular matrices reveal new facets of hepatocellular biology in culture and may be more broadly useful in culture of other soft tissues.
Collapse
Affiliation(s)
- Geeta Mehta
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
| | - Courtney M. Williams
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
| | - Luis Alvarez
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
| | - Martha Lesniewski
- Department of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Roger D. Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
| | - Linda G. Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
- Center for Gynepathology Research and Center for Environmental Health Sciences, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-429, Cambridge, MA 02139, USA
| |
Collapse
|
506
|
Kopesky PW, Lee HY, Vanderploeg EJ, Kisiday JD, Frisbie DD, Plaas AHK, Ortiz C, Grodzinsky AJ. Adult equine bone marrow stromal cells produce a cartilage-like ECM mechanically superior to animal-matched adult chondrocytes. Matrix Biol 2010; 29:427-38. [PMID: 20153827 PMCID: PMC2894996 DOI: 10.1016/j.matbio.2010.02.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/03/2010] [Accepted: 02/03/2010] [Indexed: 11/26/2022]
Abstract
Our objective was to evaluate the age-dependent mechanical phenotype of bone marrow stromal cell- (BMSC-) and chondrocyte-produced cartilage-like neo-tissue and to elucidate the matrix-associated mechanisms which generate this phenotype. Cells from both immature (2-4 month-old foals) and skeletally-mature (2-5 year-old adults) mixed-breed horses were isolated from animal-matched bone marrow and cartilage tissue, encapsulated in self-assembling-peptide hydrogels, and cultured with and without TGF-beta1 supplementation. BMSCs and chondrocytes from both donor ages were encapsulated with high viability. BMSCs from both ages produced neo-tissue with higher mechanical stiffness than that produced by either young or adult chondrocytes. Young, but not adult, chondrocytes proliferated in response to TGF-beta1 while BMSCs from both age groups proliferated with TGF-beta1. Young chondrocytes stimulated by TGF-beta1 accumulated ECM with 10-fold higher sulfated-glycosaminoglycan content than adult chondrocytes and 2-3-fold higher than BMSCs of either age. The opposite trend was observed for hydroxyproline content, with BMSCs accumulating 2-3-fold more than chondrocytes, independent of age. Size-exclusion chromatography of extracted proteoglycans showed that an aggrecan-like peak was the predominant sulfated proteoglycan for all cell types. Direct measurement of aggrecan core protein length and chondroitin sulfate chain length by single molecule atomic force microscopy imaging revealed that, independent of age, BMSCs produced longer core protein and longer chondroitin sulfate chains, and fewer short core protein molecules than chondrocytes, suggesting that the BMSC-produced aggrecan has a phenotype more characteristic of young tissue than chondrocyte-produced aggrecan. Aggrecan ultrastructure, ECM composition, and cellular proliferation combine to suggest a mechanism by which BMSCs produce a superior cartilage-like neo-tissue than either young or adult chondrocytes.
Collapse
Affiliation(s)
- P W Kopesky
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
507
|
Sun J, Zheng Q, Wu Y, Liu Y, Guo X, Wu W. Biocompatibility of KLD-12 peptide hydrogel as a scaffold in tissue engineering of intervertebral discs in rabbits. ACTA ACUST UNITED AC 2010; 30:173-7. [DOI: 10.1007/s11596-010-0208-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Indexed: 01/08/2023]
|
508
|
Park JS, Woo DG, Yang HN, Lim HJ, Park KM, Na K, Park KH. Chondrogenesis of human mesenchymal stem cells encapsulated in a hydrogel construct: neocartilage formation in animal models as both mice and rabbits. J Biomed Mater Res A 2010; 92:988-96. [PMID: 19296541 DOI: 10.1002/jbm.a.32341] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study, in vivo studies, both nude mouse and rabbit cartilage defect, were tested for chondrogenesis using stem cells (SCs) using growth factor. Specifically, human mesenchymal stem cells (hMSCs) were embedded in a hydrogel scaffold, which was coencapsulated with transforming growth factor-beta3 (TGF-beta3). The specific extracellular matrices (ECMs) released from hMSCs transplanted into the animal were assessed via glycosaminoglycan (GAG)/DNA content, RT-PCR, real time-QPCR, immunohistochemical (IHC), and Safranin-O staining and were observed up to 7 weeks after injection. By detection of ECMs the GAG content per cell remained constant for all formulations, indicating that the dramatic increase in cell number for samples with TGF-beta3 was accompanied by the maintenance of the cell phenotypes. The histological and IHC staining of the newly repaired tissues observed after treatment with TGF-beta3 mixed with hMSCs evidenced hyaline cartilage-like characteristics. Moreover, the results observed with the animal model (rabbit) treated with hMSCs embedded in the growth factor-containing hydrogel indicate that the implantation of mixed cells with TGF-beta3 may constitute a clinically efficient method for the regeneration of hyaline articular cartilage.
Collapse
Affiliation(s)
- Ji Sun Park
- College of Medicine, Pochon CHA University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
509
|
Wang Y, Zhao L, Hantash BM. Support of human adipose-derived mesenchymal stem cell multipotency by a poloxamer-octapeptide hybrid hydrogel. Biomaterials 2010; 31:5122-30. [PMID: 20347134 DOI: 10.1016/j.biomaterials.2010.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 03/03/2010] [Indexed: 11/19/2022]
Abstract
The development of new biological materials, particularly those capable of serving as permissive substrates for cell growth, differentiation, and biological function, is a key area for advancing medical technology. In this work, we examined the characteristics of a hybrid hydrogel scaffold composed of poloxamer 407 (PO) and the self-assembling oligopeptdide EFK8 in vitro and in vivo. Rheological tests showed that the storage modulus of EFK8-PO increased by 4 orders of magnitude compared to that of EFK8 alone, indicating that EFK8-PO integrates PO's high and tunable mechanical strength and integrity with the superior bioactivity of EFK8. When human adipose-derived mesenchymal stem cells (hAMSCs) were cultured in PO, we observed severe aggregation. Conversely, almost no aggregation was observed in EFK8 or EFK8-PO after 6 days of culture. hAMSC viability in all 3 hydrogels remained above 80% after 2 weeks of culture. EFK8 and EFK8-PO significantly increased hAMSC proliferation rates. In addition, EFK8- and EFK8-PO- but not PO encapsulated hAMSCs differentiated into adipocytes or osteoblasts when exposed to appropriate induction medium, suggesting EFK8 supports hAMSC multipotency in vitro. Moreover, only EFK8-PO supported hAMSC engraftment and adipogenic differenitiation post-transplantation into nude mice. Immunohistochemical analysis confirmed the new tissue was human in origin. Our studies show that EFK8-PO maintains improved mechanical properties and bioactivity relative to its individual constituents, supporting its potential use as a stem cell scaffold in soft tissue engineering.
Collapse
Affiliation(s)
- Ying Wang
- Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine, 257 Campus Dr, Stanford, CA 94305, USA
| | | | | |
Collapse
|
510
|
Nakamura M, Iwanaga S, Henmi C, Arai K, Nishiyama Y. Biomatrices and biomaterials for future developments of bioprinting and biofabrication. Biofabrication 2010; 2:014110. [PMID: 20811125 DOI: 10.1088/1758-5082/2/1/014110] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The next step beyond conventional scaffold-based tissue engineering is cell-based direct biofabrication techniques. In industrial processes, various three-dimensional (3D) prototype models have been fabricated using several different rapid prototyping methods, such as stereo-lithography, 3D printing and laser sintering, as well as others, in which a variety of chemical materials are utilized. However, with direct cell-based biofabrication, only biocompatible materials can be used, and the manufacturing process must be performed under biocompatible and physiological conditions. We have developed a direct 3D cell printing system using inkjet and gelation techniques with inkjet droplets, and found that it had good potential to construct 3D structures with multiple types of cells. With this system, we have used alginate and fibrin hydrogel materials, each of which has advantages and disadvantages. Herein, we discuss the roles of hydrogel for biofabrication and show that further developments in biofabrication technology with biomatrices will play a major part, as will developments in manufacturing technology. It is important to explore suitable biomatrices as the next key step in biofabrication techniques.
Collapse
Affiliation(s)
- M Nakamura
- Graduate School of Science and Engineering for Research, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan.
| | | | | | | | | |
Collapse
|
511
|
Maher SA, Mauck RL, Rackwitz L, Tuan RS. A nanofibrous cell-seeded hydrogel promotes integration in a cartilage gap model. J Tissue Eng Regen Med 2010; 4:25-9. [PMID: 19834956 DOI: 10.1002/term.205] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The presence of a defect in mature articular cartilage can lead to degenerative changes of the joint. This is in part caused by the inability of cartilage to regenerate tissue that is capable of spanning a fissure or crack. In this study, we hypothesized that introduction of a biodegradable cell-seeded nanofibrous hydrogel, Puramatrix(), into a cartilage gap would facilitate the generation of a mechanically stable interface. The effects of chondrocyte incorporation within the hydrogel and supplementation with transforming growth factor-beta3 (TGFbeta3), a known regulator of cell growth and differentiation, on cartilage integration were examined mechanically and histologically as a function of cell density and incubation time. When supplemented with TGFbeta3, the cell-seeded hydrogel exhibited abundant matrix generation within the hydrogel and a corresponding increase in maximum push-out stress as compared to all other groups. Furthermore, initial cell seeding density affected interfacial strength in a time-dependent manner. This study suggests that a cell-seeded TGFbeta3-supplemented hydrogel can encourage integration between two opposing pieces of articular cartilage.
Collapse
Affiliation(s)
- S A Maher
- Hospital for Special Surgery, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
512
|
Kim M, Shin Y, Hong BH, Kim YJ, Chun JS, Tae G, Kim YH. In Vitro Chondrocyte Culture in a Heparin-Based Hydrogel for Cartilage Regeneration. Tissue Eng Part C Methods 2010; 16:1-10. [DOI: 10.1089/ten.tec.2008.0548] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mihye Kim
- Research Center for Biomolecular Nanotechnology, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Youngnim Shin
- Research Center for Biomolecular Nanotechnology, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Bo-Hee Hong
- Research Center for Biomolecular Nanotechnology, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yang-Jung Kim
- Department of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Jang-Soo Chun
- Research Center for Biomolecular Nanotechnology, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Giyoong Tae
- Research Center for Biomolecular Nanotechnology, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Young Ha Kim
- Research Center for Biomolecular Nanotechnology, Gwangju Institute of Science and Technology, Gwangju, Korea
- Department of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
513
|
Dutt A, Spencer E, Howard J, Pramanik A. Studies of Amyloid-Like Fibrillogenesis throughβ-Sheet-Mediated Self-Assembly of Short Synthetic Peptides. Chem Biodivers 2010; 7:363-75. [DOI: 10.1002/cbdv.200900050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
514
|
Chai D, Arner E, Griggs D, Grodzinsky A. Alphav and beta1 integrins regulate dynamic compression-induced proteoglycan synthesis in 3D gel culture by distinct complementary pathways. Osteoarthritis Cartilage 2010; 18:249-56. [PMID: 19800448 PMCID: PMC2818306 DOI: 10.1016/j.joca.2009.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 08/25/2009] [Accepted: 09/09/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Our goal was to test the hypothesis that specific integrin receptors regulate chondrocyte biosynthetic response to dynamic compression at early times in 3D gel culture, during initial evolution of the pericellular matrix, but prior to significant accumulation of further-removed matrix. The study was motivated by increased use of dynamic loading, in vitro, for early stimulation of tissue engineered cartilage, and the need to understand the effects of loading, in vivo, at early times after implantation of constructs. METHODS Bovine articular chondrocytes were seeded in 2% agarose gels (15x10(6)cells/mL) and incubated for 18 h with and without the presence of specific integrin blockers (small-molecule peptidomimetics, function-blocking antibodies, and RGD-containing disintegrins). Samples were then subjected to a 24-h dynamic compression regime found previously to stimulate chondrocyte biosynthesis in 3D gel as well as cartilage explant culture (1 Hz, 2.5% dynamic strain amplitude, 7% static offset strain). At the end of loading, proteoglycan (PG) synthesis ((35)S-sulfate incorporation), protein synthesis ((3)H-proline incorporation), DNA content (Hoechst dye 33258) and total glycosaminoglycan (GAG) content (dimethyl methylene blue (DMMB) dye binding) were assessed. RESULTS Consistent with previous studies, dynamic compression increased PG synthesis and total GAG accumulation compared to free-swelling controls. Blocking alphavbeta3 abolished this response, independent of effects on controls, while blocking beta1 abolished the relative changes in synthesis when changes in free-swelling synthesis rates were observed. CONCLUSIONS This study suggests that both alphavbeta3 and beta1 play a role in pathways that regulate stimulation of PG synthesis and accumulation by dynamic compression, but through distinct complementary mechanisms.
Collapse
Affiliation(s)
- D.H. Chai
- Biological Engineering Department, Massachusetts Institute of Technology
| | | | | | - A.J. Grodzinsky
- Biological Engineering Department, Massachusetts Institute of Technology, Electrical, Mechanical Engineering Departments, Massachusetts Institute of Technology
| |
Collapse
|
515
|
Kopesky PW, Vanderploeg EJ, Sandy JS, Kurz B, Grodzinsky AJ. Self-assembling peptide hydrogels modulate in vitro chondrogenesis of bovine bone marrow stromal cells. Tissue Eng Part A 2010; 16:465-77. [PMID: 19705959 PMCID: PMC2862611 DOI: 10.1089/ten.tea.2009.0158] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2009] [Accepted: 08/24/2009] [Indexed: 11/13/2022] Open
Abstract
Our objective was to test the hypothesis that self-assembling peptide hydrogel scaffolds provide cues that enhance the chondrogenic differentiation of bone marrow stromal cells (BMSCs). BMSCs were encapsulated within two unique peptide hydrogel sequences, and chondrogenesis was compared with that in agarose hydrogels. BMSCs in all three hydrogels underwent transforming growth factor-beta1-mediated chondrogenesis as demonstrated by comparable gene expression and biosynthesis of extracellular matrix molecules. Expression of an osteogenic marker was unchanged, and an adipogenic marker was suppressed by transforming growth factor-beta1 in all hydrogels. Cell proliferation occurred only in the peptide hydrogels, not in agarose, resulting in higher glycosaminoglycan content and more spatially uniform proteoglycan and collagen type II deposition. The G1-positive aggrecan produced in peptide hydrogels was predominantly the full-length species, whereas that in agarose was predominantly the aggrecanase product G1-NITEGE. Unique cell morphologies were observed for BMSCs in each peptide hydrogel sequence, with extensive cell-cell contact present for both, whereas BMSCs in agarose remained rounded over 21 days in culture. Differences in cell morphology within the two peptide scaffolds may be related to sequence-specific cell adhesion. Taken together, this study demonstrates that self-assembling peptide hydrogels enhance chondrogenesis compared with agarose as shown by extracellular matrix production, DNA content, and aggrecan molecular structure.
Collapse
Affiliation(s)
- Paul W. Kopesky
- Department of Biological Engineering, MIT, Cambridge, Massachusetts
| | | | - John S. Sandy
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois
| | | | | |
Collapse
|
516
|
Self-assembling peptide nanofiber scaffolds, platelet-rich plasma, and mesenchymal stem cells for injectable bone regeneration with tissue engineering. J Craniofac Surg 2010; 20:1523-30. [PMID: 19816290 DOI: 10.1097/scs.0b013e3181b09b7e] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The purpose of this study was to investigate a capability of PuraMatrix (PM), which is a self-assembling peptide nanomaterial, as a scaffold for bone regeneration in combination with dog mesenchymal stem cells (dMSCs) and/or platelet-rich plasma (PRP) using tissue engineering and regenerative technology. Initially, teeth were extracted from an adult hybrid dog's mandible region. After 4 weeks, bone defects were prepared on both sides of the mandible with a trephine bar. The following graft materials were implanted into these defects: (1) control (defect only), (2) PM, (3) PM/PRP, (4) PM/dMSCs, and (5) PM/dMSCs/PRP. From scanning electron microscope images, PM had a three-dimensional nanostructure, and dMSCs attached on the surface of PM. At 2, 4, and 8 weeks after implantation, each sample was collected from the graft area with a trephine bar and assessed by histologic and histomorphometric analyses. It was observed that the bone regenerated by PM/dMSCs/PRP was of excellent quality, and mature bone had been formed. Histometrically, at 8 weeks, newly formed bone areas comprised 12.39 +/- 1.29% (control), 25.28 +/- 3.92% (PM), 27.72 +/- 3.15% (PM/PRP), 50.07 +/- 3.97% (PM/dMSCs), and 58.43 +/- 5.06% (PM/dMSCs/PRP). The PM/dMSCs and PM/dMSCs/PRP groups showed a significant increase at all weeks compared with the control, PM, or PM/PRP (P < 0.05 at 2, 4, and 8 weeks, analysis of variance). These results showed that MSCs might keep their own potential and promote new bone regeneration in the three-dimensional structure by PM scaffolds. Taken together, it is suggested that PM might be useful as a scaffold of bone regeneration in cell therapy, and these results might lead to an effective treatment method for bone defects.
Collapse
|
517
|
Ozeki M, Kuroda S, Kon K, Kasugai S. Differentiation of bone marrow stromal cells into osteoblasts in a self-assembling peptide hydrogel: in vitro and in vivo studies. J Biomater Appl 2010; 25:663-84. [PMID: 20089608 DOI: 10.1177/0885328209356328] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
A prerequisite of tissue engineering approaches with regard to autograft is a suitable scaffold that can harbor cells and signals. Conventionally, such scaffolds have been prepared as 3D scaffolds prefabricated from synthetic or natural biomaterials. RAD16 has been introduced as a new biomaterial, where synthetic peptides self-assemble to form a hydrogel. In this study, RAD16 was examined in terms of osteogenic efficacy and feasibility of ectopic mineralization. Two hundred and seventy-one RAD16 was cocultured with 1 × 10(6) bone marrow cells from the femurs of 6-week-old Wistar male rats in alpha minimum essential medium supplemented with or without dexamethasone. Second, the same volume of the RAD16 construct hosting the cells with or without hydroxyapatite (HA) particles was treated in the dexamethasone medium as well, prepared in a Teflon tube, and implanted subcutaneously. Cell proliferation was prominent in the RAD16 coculture with dexamethasone at 1 week and significantly decreased by 2 weeks, whereas the other combinations remained or inclined, and their osteogenic differentiation was accelerated up to 2 weeks, as seen in increasing alkaline phosphatase (ALP) activity and mRNAs of ALP, OPN, and OCN. The RAD16 implant prepared with HA particles allowed more osteoblast-like cells and blood cells to grow inside, which was accompanied by elevating OPN gene expression and the stronger peak of VEGF gene expression at 2 weeks. Furthermore, more OPN mRNA signal was detected around the RAD16 containing HA particles by 4 weeks. On the other hand, the RAD16 alone represented lower expression of OPN gene. During the experiment, however, no ectopic mineralization was observed in both groups. Conclusively, it was suggested that the RAD16 showed feasibility of serving as a matrix for osteogenic differentiation of cocultured bone marrow cells in vitro and in vivo. Proceeding of exploration and modification of RAD16 are continuously required for cell-based tissue engineering.
Collapse
Affiliation(s)
- Maho Ozeki
- Oral Implantology and Regenerative Dental Medicine Graduate School, Tokyo Medical and Dental University 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
518
|
Chen C, Pan F, Zhang S, Hu J, Cao M, Wang J, Xu H, Zhao X, Lu JR. Antibacterial Activities of Short Designer Peptides: a Link between Propensity for Nanostructuring and Capacity for Membrane Destabilization. Biomacromolecules 2010; 11:402-11. [PMID: 20078032 DOI: 10.1021/bm901130u] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Cuixia Chen
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Fang Pan
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Shengzhong Zhang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Jing Hu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Jing Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Hai Xu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Xiubo Zhao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| | - Jian R. Lu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao Economic Development Zone, Qingdao 266555, China, and Biological Physics Laboratory, School of Physics and Astronomy, University of Manchester, Schuster Building, Manchester M13 9PL, United Kingdom
| |
Collapse
|
519
|
Desii A, Chiellini F, Di Stefano R, Tiné MR, Solaro R. Hydrogel scaffolds by self-assembly of a complementary ionic tetrapeptide. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/pola.23841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
520
|
Gentsch R, Börner HG. Designing Three-Dimensional Materials at the Interface to Biology. BIOACTIVE SURFACES 2010. [DOI: 10.1007/12_2010_80] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
521
|
Hauser CAE, Zhang S. Designer self-assembling peptide nanofiber biological materials. Chem Soc Rev 2010; 39:2780-90. [DOI: 10.1039/b921448h] [Citation(s) in RCA: 295] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
522
|
Abstract
Peptide nanostructures containing bioactive signals offer exciting novel therapies of broad potential impact in regenerative medicine. These nanostructures can be designed through self-assembly strategies and supramolecular chemistry, and have the potential to combine bioactivity for multiple targets with biocompatibility. It is also possible to multiplex their functions by using them to deliver proteins, nucleic acids, drugs and cells. In this review, we illustrate progress made in this new field by our group and others using peptide-based nanotechnology. Specifically, we highlight the use of self-assembling peptide amphiphiles towards applications in the regeneration of the central nervous system, vasculature and hard tissue along with the transplant of islets and the controlled release of nitric oxide to prevent neointimal hyperplasia. Also, we discuss other self-assembling oligopeptide technology and the progress made with these materials towards the development of potential therapies.
Collapse
Affiliation(s)
- M J Webber
- Northwestern University Department of Biomedical Engineering, Evanston, IL, USA
| | | | | |
Collapse
|
523
|
Cavalli S, Albericio F, Kros A. Amphiphilic peptides and their cross-disciplinary role as building blocks for nanoscience. Chem Soc Rev 2010; 39:241-63. [DOI: 10.1039/b906701a] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
524
|
Abstract
This review presents an overview on bio-hybrid approaches of integrating the structural and functional features of proteins and peptides with synthetic polymers and the resulting unique properties in such hybrids, with a focus on bioresponsive/bioactive systems with biomaterials applications. The review is divided in two broad sections. First, we describe several examples of bio-hybrids produced by combining versatile synthetic polymers with proteins/enzymes and drugs that have resulted in (1) hybrid materials based on responsive polymers, (2) responsive hydrogels based on enzyme-catalyzed reactions, protein-protein interactions and protein-drug sensing, and (3) dynamic hydrogels based on conformational changes of a protein. Next, we present hybrids produced by combining synthetic polymers with peptides, classified based on the properties of the peptide domain: (1) peptides with different conformations, such as alpha-helical, coiled-coil, and beta-sheet; (2) peptides derived from structural protein domains such as silk, elastin, titin, and collagen; and (3) peptides with other biofunctional properties such as cell-binding domains and enzyme-recognized degradation domains.
Collapse
Affiliation(s)
- Ohm D. Krishna
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711
| | - Kristi L. Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711
| |
Collapse
|
525
|
Rajagopal K, Lamm MS, Haines-Butterick LA, Pochan DJ, Schneider JP. Tuning the pH responsiveness of beta-hairpin peptide folding, self-assembly, and hydrogel material formation. Biomacromolecules 2009; 10:2619-25. [PMID: 19663418 DOI: 10.1021/bm900544e] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A design strategy to control the thermally triggered folding, self-assembly, and subsequent hydrogelation of amphiphilic beta-hairpin peptides in a pH-dependent manner is presented. Point substitutions of the lysine residues of the self-assembling peptide MAX1 were made to alter the net charge of the peptide. In turn, the electrostatic nature of the peptide directly influences the solution pH at which thermally triggered hydrogelation is permitted. CD spectroscopy and oscillatory rheology show that peptides of lower net positive charge are capable of folding and assembling into hydrogel material at lower values of pH at a given temperature. The pH sensitive folding and assembling behavior is not only dependent on the net peptide charge, but also on the exact position of substitution within the peptide sequence. TEM shows that these peptides self-assemble into hydrogels that are composed of well-defined fibrils with nonlaminated morphologies. TEM also indicates that fibril morphology is not influenced by making these sequence changes on the hydrophilic face of the hairpins. Rheology shows that the ultimate mechanical rigidity of these peptide hydrogels is dependent on the rate of folding and self-assembly. Peptides that fold and assemble faster afford more rigid gels. Ultimately, this design strategy yielded a peptide MAX1(K15E) that is capable of undergoing thermally triggered hydrogelation at physiological buffer conditions (pH 7.4, 150 NaCl, 37 degrees C).
Collapse
Affiliation(s)
- Karthikan Rajagopal
- Department of Chemistry and Biochemistry, University of Delaware, Newark Delaware 19716, USA
| | | | | | | | | |
Collapse
|
526
|
Orbach R, Adler-Abramovich L, Zigerson S, Mironi-Harpaz I, Seliktar D, Gazit E. Self-assembled Fmoc-peptides as a platform for the formation of nanostructures and hydrogels. Biomacromolecules 2009; 10:2646-51. [PMID: 19705843 DOI: 10.1021/bm900584m] [Citation(s) in RCA: 244] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hydrogels are of great interest as a class of materials for tissue engineering, axonal regeneration, and controlled drug delivery, as they offer 3D interwoven scaffolds to support the growth of cells. Herein, we extend the family of the aromatic Fmoc-dipeptides with a library of new Fmoc-peptides, which include natural and synthetic amino acids with an aromatic nature. We describe the self-assembly of these Fmoc-peptides into various structures and characterize their distinctive molecular and physical properties. Moreover, we describe the fabrication of the bioactive RGD sequence into a hydrogel. This unique material offers new opportunities for developing cell-adhesive biomedical hydrogel scaffolds, as well as for establishing strategies to modify surfaces with bioactive materials.
Collapse
Affiliation(s)
- Ron Orbach
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6407536, Israel
| | | | | | | | | | | |
Collapse
|
527
|
Kisiday JD, Frisbie DD, McIlwraith CW, Grodzinsky AJ. Dynamic compression stimulates proteoglycan synthesis by mesenchymal stem cells in the absence of chondrogenic cytokines. Tissue Eng Part A 2009; 15:2817-24. [PMID: 19243241 DOI: 10.1089/ten.tea.2008.0357] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The objective of this study was to evaluate the effect of dynamic compression on mesenchymal stem cell (MSC) chondrogenesis. Dynamic compression was applied to agarose hydrogels seeded with bone marrow-derived adult equine MSCs. In the absence of the chondrogenic cytokine transforming growth factor beta (TGFbeta), dynamic compression applied for 12 h per day led to significantly greater proteoglycan synthesis than in unloaded TGFbeta-free cultures, although at a rate that was approximately 20% to 35% of unloaded TGFbeta cultures. These data suggest that the emergence of aggrecan dominated a chondrogenic response to loading as increases in proteoglycan synthesis. Cross-sectional analyses were conducted to subjectively identify potential spatial distributions of heterogeneous differentiation. In loaded samples, cell viability and metachromatic staining was low near the porous compression platen interface but increased with depth, reaching levels in the lower portion of the hydrogel that resembled unloaded TGFbeta cultures. These results suggest that the combination of high hydrostatic pressure and low dynamic strain and fluid flow had a stronger effect on chondrogenesis than did low hydrostatic pressure coupled with high dynamic strain and fluid flow. Next, the 12-h per day loading protocol was applied in the presence of TGFbeta. Biosynthesis in loaded cultures was less than in unloaded TGFbeta samples. Taken together, these data suggest that the duration of loading necessary to stimulate mechanoinduction of MSCs may not be optimal for neo-tissue accumulation in the presence of chondrogenic cytokines.
Collapse
Affiliation(s)
- John D Kisiday
- Orthopaedic Research Center, Department of Clinical Science, Colorado State University, Fort Collins, Colorado 80523, USA.
| | | | | | | |
Collapse
|
528
|
Nakatani S, Mano H, Sampei C, Shimizu J, Wada M. Chondroprotective effect of the bioactive peptide prolyl-hydroxyproline in mouse articular cartilage in vitro and in vivo. Osteoarthritis Cartilage 2009; 17:1620-7. [PMID: 19615963 DOI: 10.1016/j.joca.2009.07.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 06/19/2009] [Accepted: 07/02/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the direct effect of prolyl-hydroxyproline (Pro-Hyp) on chondrocytes under in vivo and in vitro conditions in an attempt to identify Pro-Hyp as the bioactive peptide in collagen hydrolysate (CH). METHODS The in vivo effects of CH and Pro-Hyp intake on articular cartilage were studied by microscopic examination of sections of dissected articular cartilage from treated C57BL/6J mice. In this study, mice that were fed diets containing excess phosphorus were used as an in vivo model. This mouse line showed loss of chondrocytes and reduced thickness of articular cartilage, with abnormality of the subchondral bone. The in vitro effects of CH, Pro-Hyp, amino acids and other peptides on proliferation, differentiation, glycosaminoglycan content and mineralization of chondrocytes were determined by MTT activity and staining with alkaline phosphatase, alcian blue and alizarin red. Expression of chondrogenesis-specific genes in ATDC5 cells was determined by semiquantitative Reverse Transcription Polymerase Chain Reaction (RT-PCR). RESULTS In vivo, CH and Pro-Hyp inhibited the loss of chondrocytes and thinning of the articular cartilage layer caused by phosphorus-induced degradation. In the in vitro study, CH and Pro-Hyp did not affect chondrocyte proliferation but inhibited their differentiation into mineralized chondrocytes. A combination of amino acids such as proline, hydroxyproline and prolyl-hydroxyprolyl-glycine did not affect chondrocyte proliferation or differentiation. Moreover, CH and Pro-Hyp caused two and threefold increases, respectively, in the staining area of glycosaminoglycan in the extracellular matrix of ATDC5 cells. RT-PCR indicated that Pro-Hyp increased the aggrecan mRNA level approximately twofold and decreased the Runx1 and osteocalcin mRNA levels by two-thirds and one-tenth, respectively. CONCLUSION Pro-Hyp is the first bioactive edible peptide derived from CH to be shown to affect chondrocyte differentiation under pathological conditions.
Collapse
Affiliation(s)
- S Nakatani
- Department of Food Functional Science, Graduate School of Pharmacology, Josai University, Keyakidai 1-1, Sakado, Saitama 3500295, Japan
| | | | | | | | | |
Collapse
|
529
|
Molecular Design of Biological and Nano-Materials. Biomimetics (Basel) 2009. [DOI: 10.1201/9781420037715.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] Open
|
530
|
Huang AH, Stein A, Tuan RS, Mauck RL. Transient exposure to transforming growth factor beta 3 improves the mechanical properties of mesenchymal stem cell-laden cartilage constructs in a density-dependent manner. Tissue Eng Part A 2009; 15:3461-72. [PMID: 19432533 PMCID: PMC2792068 DOI: 10.1089/ten.tea.2009.0198] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 05/11/2009] [Indexed: 01/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive cell source for cartilage tissue engineering and regenerative medicine. However, the use of these cells has been limited by their reduced ability to form functional tissue compared to chondrocytes when placed in three-dimensional culture systems. To optimize MSC functional chondrogenesis, we examined the effects of increasing seeding density and transient application of transforming growth factor beta 3 (TGF-beta3), two factors previously shown to improve growth of chondrocyte-based constructs. Chondrocytes seeded in agarose at 20 million cells/mL and MSCs seeded at 20 or 60 million cells/mL agarose were cultured for 7 weeks under continuous or transient application of TGF-beta3. In the transient group, cell-laden constructs were exposed to TGF-beta3 for the initial 3 weeks, followed by 4 weeks of culture in medium without TGF-beta3. Compressive properties, biochemical content, and gene expression were assessed at 3, 5, and 7 weeks. Matrix distribution and collagen type was determined using histology and immunohistochemistry, and chondrogenic and osteogenic markers were assessed using real-time polymerase chain reaction. When maintained continuously with TGF-beta3, chondrocyte-seeded constructs achieved a higher equilibrium compressive modulus than MSCs similarly maintained. Although properties of both groups increased with respect to starting values, there was no difference in bulk mechanical or biochemical properties with higher seeding density when MSCs were cultured with constant TGF-beta3. Findings also showed that while transient application of TGF-beta3 elicited robust growth from chondrocyte-laden gels, MSCs seeded at the same density failed to respond, although constructs maintained their previously accrued properties and continued to express cartilaginous genes after TGF-beta3 removal. Conversely, MSCs seeded at 60 million cells/mL exhibited a strong anabolic response with transient TGF-beta3 exposure, achieving an equilibrium modulus of approximately 200 kPa. Although this represents the highest modulus we have been able to achieve with MSC-seeded constructs using our culture system, further work remains to optimize MSC chondrogenesis for cartilage tissue engineering, particularly in terms of collagen content and dynamic mechanical properties.
Collapse
Affiliation(s)
- Alice H. Huang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ashley Stein
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
531
|
Zhang H, Luo H, Zhao X. Mechanistic Study of Self-Assembling Peptide RADA16-I in Formation of Nanofibers and Hydrogels. J Nanotechnol Eng Med 2009. [DOI: 10.1115/1.4000301] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The biophysical and biochemical properties of RADA16-I, the representative of a class of self-assembling peptides, were studied to elucidate the molecular mechanism of nanofiber and hydrogel formations. We found that self-assembly occurs in the solution at low pH (pH 4), rather than the popular belief that it occurs in the physiological environment. Actually, the peptide lost its β-sheet structure and formed irregular aggregates in the condition around pH 7. Our results demonstrated that the extended conformation of peptide backbone caused by the electrostatic repulsive force in acid solution is crucial for the peptide to self-assemble into nanofibers. Importantly, we have proposed a mechanism for the peptide to form nanofiber hydrogel in the physiological condition, which is not propitious for nanofiber formation. Hypothetically, it is by virtue of the tendency of fibers to collapse and form irregular aggregates at pH 7 that we could obtain stable hydrogels by introducing phosphate buffered saline into the system.
Collapse
Affiliation(s)
- Hangyu Zhang
- Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, No.1, Ke Yuan 4th Street, Gao Peng Road, Chengdu, 610041 Sichuan, China
| | - Hanlin Luo
- Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, No.1, Ke Yuan 4th Street, Gao Peng Road, Chengdu, 610041 Sichuan, China
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, No.1, Ke Yuan 4th Street, Gao Peng Road, Chengdu, 610041 Sichuan, China; Center for Biomedical Engineering, NE47-378, Massachusetts Institute of Technology, Cambridge, MA 02139-4307
| |
Collapse
|
532
|
Sallach RE, Cui W, Balderrama F, Martinez AW, Wen J, Haller CA, Taylor JV, Wright ER, Long RC, Chaikof EL. Long-term biostability of self-assembling protein polymers in the absence of covalent crosslinking. Biomaterials 2009; 31:779-91. [PMID: 19854505 DOI: 10.1016/j.biomaterials.2009.09.082] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Accepted: 09/21/2009] [Indexed: 11/29/2022]
Abstract
Unless chemically crosslinked, matrix proteins, such as collagen or silk, display a limited lifetime in vivo with significant degradation observed over a period of weeks. Likewise, amphiphilic peptides, lipopeptides, or glycolipids that self-assemble through hydrophobic interactions to form thin films, fiber networks, or vesicles do not demonstrate in vivo biostability beyond a few days. We report herein that a self-assembling, recombinant elastin-mimetic triblock copolymer elicited minimal inflammatory response and displayed robust in vivo stability for periods exceeding 1 year, in the absence of either chemical or ionic crosslinking. Specifically, neither a significant inflammatory response nor calcification was observed upon implantation of test materials into the peritoneal cavity or subcutaneous space of a mouse model. Moreover, serial quantitative magnetic resonance imaging, evaluation of pre- and post-explant ultrastructure by cryo-high resolution scanning electron microscopy, and an examination of implant mechanical responses revealed substantial preservation of form, material architecture, and biomechanical properties, providing convincing evidence of a non-chemically or ionically crosslinked protein polymer system that exhibits long-term stability in vivo.
Collapse
Affiliation(s)
- Rory E Sallach
- Department of Surgery, Emory University, Atlanta, GA 30332, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
533
|
Telomerase activity and hepatic functions of rat embryonic liver progenitor cell in nanoscaffold-coated model bioreactor. Mol Cell Biochem 2009; 336:137-49. [DOI: 10.1007/s11010-009-0266-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
|
534
|
The use of green fluorescence gene (GFP)-modified rabbit mesenchymal stem cells (rMSCs) co-cultured with chondrocytes in hydrogel constructs to reveal the chondrogenesis of MSCs. Biomaterials 2009; 30:6374-85. [DOI: 10.1016/j.biomaterials.2009.07.062] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2009] [Accepted: 07/28/2009] [Indexed: 01/22/2023]
|
535
|
Jung Y, Kim SH, Kim YH, Kim SH. The effects of dynamic and three-dimensional environments on chondrogenic differentiation of bone marrow stromal cells. Biomed Mater 2009; 4:055009. [DOI: 10.1088/1748-6041/4/5/055009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
536
|
Slaughter BV, Khurshid SS, Fisher OZ, Khademhosseini A, Peppas NA. Hydrogels in regenerative medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2009; 21:3307-29. [PMID: 20882499 PMCID: PMC4494665 DOI: 10.1002/adma.200802106] [Citation(s) in RCA: 1832] [Impact Index Per Article: 114.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Hydrogels, due to their unique biocompatibility, flexible methods of synthesis, range of constituents, and desirable physical characteristics, have been the material of choice for many applications in regenerative medicine. They can serve as scaffolds that provide structural integrity to tissue constructs, control drug and protein delivery to tissues and cultures, and serve as adhesives or barriers between tissue and material surfaces. In this work, the properties of hydrogels that are important for tissue engineering applications and the inherent material design constraints and challenges are discussed. Recent research involving several different hydrogels polymerized from a variety of synthetic and natural monomers using typical and novel synthetic methods are highlighted. Finally, special attention is given to the microfabrication techniques that are currently resulting in important advances in the field.
Collapse
Affiliation(s)
- Brandon V. Slaughter
- Department of Biomedical Engineering, C0800, The University of Texas at
Austin, Austin, TX 78712 (USA)
| | - Shahana S. Khurshid
- Department of Biomedical Engineering, C0800, The University of Texas at
Austin, Austin, TX 78712 (USA)
| | - Omar Z. Fisher
- Department of Biomedical Engineering, C0800, The University of Texas at
Austin, Austin, TX 78712 (USA)
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and
Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health
Sciences and Technology, Massachusetts, Institute of Technology, Cambridge, MA 02139
(USA)
| | - Nicholas A. Peppas
- Biomaterials, Drug Delivery, Bionanotechnology, and Molecular, Recognition
Laboratories, Department of Chemical Engineering, C0400, The University of Texas at
Austin, Austin, TX 78712 (USA)
- Department of Pharmaceutics, C0400, The University of Texas at Austin,
Austin, TX 78712 (USA)
- Department of Biomedical Engineering, C0800, The University of Texas at
Austin, Austin, TX 78712 (USA)
| |
Collapse
|
537
|
Weinand C, Gupta R, Weinberg E, Madisch I, Neville CM, Jupiter JB, Vacanti JP. Toward Regenerating a Human ThumbIn Situ. Tissue Eng Part A 2009; 15:2605-15. [DOI: 10.1089/ten.tea.2008.0467] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Christian Weinand
- Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Rajiv Gupta
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Eli Weinberg
- Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ijad Madisch
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Craig M. Neville
- Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Jesse B. Jupiter
- Hand and Upper Extremity Service, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Joseph P. Vacanti
- Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
538
|
Kao B, Kadomatsu K, Hosaka Y. Construction of Synthetic Dermis and Skin Based on a Self-Assembled Peptide Hydrogel Scaffold. Tissue Eng Part A 2009; 15:2385-96. [DOI: 10.1089/ten.tea.2008.0362] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Bunsho Kao
- Department of Plastic and Reconstructive Surgery, School of Medicine, Showa University, Tokyo, Japan
| | - Koichi Kadomatsu
- Department of Plastic and Reconstructive Surgery, School of Medicine, Showa University, Tokyo, Japan
| | - Yoshiaki Hosaka
- Department of Plastic and Reconstructive Surgery, School of Medicine, Showa University, Tokyo, Japan
| |
Collapse
|
539
|
Nicodemus GD, Bryant SJ. Cell encapsulation in biodegradable hydrogels for tissue engineering applications. TISSUE ENGINEERING PART B-REVIEWS 2009; 14:149-65. [PMID: 18498217 DOI: 10.1089/ten.teb.2007.0332] [Citation(s) in RCA: 794] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Encapsulating cells in biodegradable hydrogels offers numerous attractive features for tissue engineering, including ease of handling, a highly hydrated tissue-like environment for cell and tissue growth, and the ability to form in vivo. Many properties important to the design of a hydrogel scaffold, such as swelling, mechanical properties, degradation, and diffusion, are closely linked to the crosslinked structure of the hydrogel, which is controlled through a variety of different processing conditions. Degradation may be tuned by incorporating hydrolytically or enzymatically labile segments into the hydrogel or by using natural biopolymers that are susceptible to enzymatic degradation. Because cells are present during the gelation process, the number of suitable chemistries and formulations are limited. In this review, we describe important considerations for designing biodegradable hydrogels for cell encapsulation and highlight recent advances in material design and their applications in tissue engineering.
Collapse
Affiliation(s)
- Garret D Nicodemus
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 80309-0424, USA
| | | |
Collapse
|
540
|
Experimental study on self-assembly of KLD-12 peptide hydrogel and 3-D culture of MSC encapsulated within hydrogel in vitro. ACTA ACUST UNITED AC 2009; 29:512-6. [DOI: 10.1007/s11596-009-0424-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Indexed: 01/08/2023]
|
541
|
Xu K, Ge W, Liang G, Wang L, Yang Z, Wang Q, Hsing IM, Xu B. Bisphosphonate-containing supramolecular hydrogels for topical decorporation of uranium-contaminated wounds in mice. Int J Radiat Biol 2009; 84:353-62. [DOI: 10.1080/09553000802029902] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
542
|
Meng H, Chen L, Ye Z, Wang S, Zhao X. The effect of a self-assembling peptide nanofiber scaffold (peptide) when used as a wound dressing for the treatment of deep second degree burns in rats. J Biomed Mater Res B Appl Biomater 2009; 89:379-391. [PMID: 18837444 DOI: 10.1002/jbm.b.31226] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
RADARADARADARADA (RADA16-I) peptide, consisting of 16 alternating hydrophobic and hydrophilic (also alternating negative and positive charges) amino acids, forms extremely stable beta-pleated sheet structure and then self-assembles into nanofibers to produce high-order interwoven nanofiber scaffold hydrogel. To investigate its therapeutic effects, a burn model of partial thickness-deep dermal injury (the deep second degree burns) was performed at the dorsal skin of female Sprague-Dawley rats with an electrical scalding machine. The wounds treated with either RADA16-I or control materials were carefully examined at morphological, histological and cellular levels. We found that RADA16-I can advance the time of eschar appearance and the time of eschar disappearance both by 3-5 days, and speed up wound contraction by 20-30% compared with contrast groups (chitosan, poly(DL)-lactic acid (PDLA), collagen I and the blank) without obvious edema. Immunohistochemical studies showed that both FGF and EGF were obviously expressed in nascent tissue such as epidermis and glands when wounds were treated with the RADA16-I after injury. When peptide stock solution was diluted from 10 to 0.17 mg/mL, atomic force microscopy (AFM) observation showed that the shape of peptide nanofibers changed from the globular-pieces-clustered filaments with 4.8 +/- 0.38 nm in height, 61.6 +/- 6.10 nm in width and 708 +/- 80.2 nm in length, to general filaments with 1.4 +/- 0.36 nm, 17.5 +/- 1.13 nm and 1108 +/- 184 nm. The nanofiber surface porosity gradually decreased from 49-70% to 12-28%. These characteristics contribute to wound healing by offering an "ideal dressing" moist healing microenvironment and a nanofiber 3D scaffold. These results suggest that the self-assembling peptide might be a promising wound dressing with being simple, effective, and affordable.
Collapse
Affiliation(s)
- Hui Meng
- Institute for NanoBiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Liyan Chen
- Institute for NanoBiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhaoyang Ye
- Institute for NanoBiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Songtao Wang
- Institute for NanoBiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaojun Zhao
- Center for Biomedical Engineering NE47-378, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
543
|
Zhang F, Shi GS, Ren LF, Hu FQ, Li SL, Xie ZJ. Designer self-assembling peptide scaffold stimulates pre-osteoblast attachment, spreading and proliferation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2009; 20:1475-1481. [PMID: 19214714 DOI: 10.1007/s10856-009-3700-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Accepted: 01/22/2009] [Indexed: 05/27/2023]
Abstract
A new peptide scaffold was made by mixing pure RADA16 (Ac-RADARADARADARADA-CONH2) and designer peptide RGDA16 (Ac-RADARGDARADARGDA-CONH2) solutions, and investigate any effect on attachment, spreading and proliferation of pre-osteoblast (MC3T3-E1). The peptides, RADA16 and RGDA16, were custom-synthesized. They were solubilized in deionized water at a concentration of 10 mg/ml (1% w/v), the RGDA16 peptide solution was mixed 1:1 with RADA16 solution and a new peptide solution RGDAmix was produced. The RGDAmix and RADA16 solution were directly loaded in 96-well plates and cover slips, and two different peptide scaffolds were formed with the addition of maintenance medium (alpha-MEM) in several minutes. About 1.0 x 10(4) MC3T3-E1 cells were seeded on each hydrogel scaffold, and then the cell morphological changes were observed using a fluorescence microscope at 1 h, 3 h and 24 h timepoint, respectively. Cell attachment was evaluated 1 h, 3 h and 24 h after cell seeding and cell proliferation was determined 4d, 7d and 14d after cell seeding. The RGDAmix scaffold significantly promoted the initial cell attachment compared with the RADA16 scaffold. MC3T3-E1 cells adhered and spread well on both scaffolds, however, cells spread better on the RGDAmix scaffold than on the RADA16 scaffold. Cell proliferation was greatly stimulated when cultured on RGDAmix scaffold. The RGD sequence contained peptide scaffold RGDAmix significantly enhances MC3T3-E1 cells attachment, spreading and proliferation.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital, College of Medicine, Zhejiang University, 395# Yan'an Road, Hangzhou 310006, People's Republic of China
| | | | | | | | | | | |
Collapse
|
544
|
Kyle S, Aggeli A, Ingham E, McPherson MJ. Production of self-assembling biomaterials for tissue engineering. Trends Biotechnol 2009; 27:423-33. [PMID: 19497631 PMCID: PMC2828541 DOI: 10.1016/j.tibtech.2009.04.002] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 04/01/2009] [Accepted: 04/01/2009] [Indexed: 11/24/2022]
Abstract
Self-assembling peptide-based biomaterials are being developed for use as 3D tissue engineering scaffolds and for therapeutic drug-release applications. Chemical synthesis provides custom-made peptides in small quantities, but production approaches based upon transgenic organisms might be more cost-effective for large-scale peptide production. Long lead times for developing appropriate animal clones or plant lines and potential negative public opinion are obstacles to these routes. Microbes, particularly safe organisms used in the food industry, offer a more rapid route to the large-scale production of recombinant self-assembling biomaterials. In this review, recent advances and challenges in the recombinant production of collagen, elastin and de novo designed self-assembling peptides are discussed.
Collapse
Affiliation(s)
- Stuart Kyle
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | | | | | | |
Collapse
|
545
|
Klein TJ, Malda J, Sah RL, Hutmacher DW. Tissue engineering of articular cartilage with biomimetic zones. TISSUE ENGINEERING. PART B, REVIEWS 2009; 15:143-57. [PMID: 19203206 PMCID: PMC3121783 DOI: 10.1089/ten.teb.2008.0563] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Accepted: 02/07/2009] [Indexed: 12/20/2022]
Abstract
Articular cartilage damage is a persistent and increasing problem with the aging population, and treatments to achieve biological repair or restoration remain a challenge. Cartilage tissue engineering approaches have been investigated for over 20 years, but have yet to achieve the consistency and effectiveness for widespread clinical use. One of the potential reasons for this is that the engineered tissues do not have or establish the normal zonal organization of cells and extracellular matrix that appears critical for normal tissue function. A number of approaches are being taken currently to engineer tissue that more closely mimics the organization of native articular cartilage. This review focuses on the zonal organization of native articular cartilage, strategies being used to develop such organization, the reorganization that occurs after culture or implantation, and future prospects for the tissue engineering of articular cartilage with biomimetic zones.
Collapse
Affiliation(s)
- Travis J. Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Jos Malda
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Robert L. Sah
- Department of Bioengineering, University of California–San Diego, La Jolla, California
| | - Dietmar W. Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
546
|
Tang C, Shao X, Sun B, Huang W, Zhao X. The effect of self-assembling peptide RADA16-I on the growth of human leukemia cells in vitro and in nude mice. Int J Mol Sci 2009; 10:2136-2145. [PMID: 19564944 PMCID: PMC2695272 DOI: 10.3390/ijms10052136] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 04/30/2009] [Accepted: 05/12/2009] [Indexed: 02/05/2023] Open
Abstract
Nanofiber scaffolds formed by self-assembling peptide RADA16-I have been used for the study of cell proliferation to mimic an extracellular matrix. In this study, we investigated the effect of RADA16-I on the growth of human leukemia cells in vitro and in nude mice. Self-assembly assessment showed that RADA16-I molecules have excellent self-assembling ability to form stable nanofibers. MTT assay displayed that RADA16-I has no cytotoxicity for leukemia cells and human umbilical vein endothelial cells (HUVECs) in vitro. However, RADA16-I inhibited the growth of K562 tumors in nude mice. Furthermore, we found RADA16-I inhibited vascular tube-formation by HUVECs in vitro. Our data suggested that nanofiber scaffolds formed by RADA16-I could change tumor microenvironments, and inhibit the growth of tumors. The study helps to encourage further design of self-assembling systems for cancer therapy.
Collapse
Affiliation(s)
- Chengkang Tang
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu, 610041, China; E-Mails:
(C.T.);
(X.S.);
(B.S.);
(W.H.)
| | - Ximing Shao
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu, 610041, China; E-Mails:
(C.T.);
(X.S.);
(B.S.);
(W.H.)
| | - Binbin Sun
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu, 610041, China; E-Mails:
(C.T.);
(X.S.);
(B.S.);
(W.H.)
| | - Wenli Huang
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu, 610041, China; E-Mails:
(C.T.);
(X.S.);
(B.S.);
(W.H.)
| | - Xiaojun Zhao
- Institute for Nanobiomedical Technology and Membrane Biology, West China Hospital, Sichuan University, Chengdu, 610041, China; E-Mails:
(C.T.);
(X.S.);
(B.S.);
(W.H.)
- West China Medical School, West China Hospital, Sichuan University, Guo Xue Xiang 37, Chengdu, 610041, Chengdu, China
- Center for Biomedical Engineering, NE47-379, Massachusetts Institute of Technology, Cambridge, MA 02139-4307, USA
- Author to whom correspondence should be addressed; E-mails:
; Tel. +86-28-8516-4069; Fax: +86-28-8516-4070
| |
Collapse
|
547
|
Mi K, Wang G, Liu Z, Feng Z, Huang B, Zhao X. Influence of a Self-Assembling Peptide, RADA16, Compared with Collagen I and Matrigel on the Malignant Phenotype of Human Breast-Cancer Cells in 3D Cultures andin vivo. Macromol Biosci 2009; 9:437-43. [DOI: 10.1002/mabi.200800262] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
548
|
Ariga K, Ji Q, Hill JP, Kawazoe N, Chen G. Supramolecular approaches to biological therapy. Expert Opin Biol Ther 2009; 9:307-20. [PMID: 19216620 DOI: 10.1517/14712590802715772] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Supramolecular chemistry is a useful methodology for construction of nano- or micro-sized objects and can significantly contribute to nanotechnology through so-called bottom-up processing. In addition, supramolecular self-assembled structures can mimic some aspects of biological systems. Bio-related functions such as molecular sensing, controlled release, signaling and materials separations have been realized. Supramolecular chemistry is a multidisciplinary field that includes subjects such as molecular design and nanosized materials. In this article recent examples of supramolecular chemistry in the context of biological therapy are introduced and classified into five categories: small supramolecular systems; designer polymers; self-assembled structures; predesigned assemblies; and nanomaterials. Finally, hierarchic organization of supramolecular structures for advanced functions is introduced to illustrate future directions of investigation. We hope that scientists studying therapeutic applications receive inspiration from this review to exploit the opportunities offered by supramolecular chemistry in their respective research areas.
Collapse
Affiliation(s)
- Katsuhiko Ariga
- National Institute for Materials Science, 1-1 Namiki, Tsukuba, 305-0044, Japan.
| | | | | | | | | |
Collapse
|
549
|
Quintana L, zur Nieden NI, Semino CE. Morphogenetic and regulatory mechanisms during developmental chondrogenesis: new paradigms for cartilage tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2009; 15:29-41. [PMID: 19063663 DOI: 10.1089/ten.teb.2008.0329] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cartilage is the first skeletal tissue to be formed during embryogenesis leading to the creation of all mature cartilages and bones, with the exception of the flat bones in the skull. Therefore, errors occurring during the process of chondrogenesis, the formation of cartilage, often lead to severe skeletal malformations such as dysplasias. There are hundreds of skeletal dysplasias, and the molecular genetic etiology of some remains more elusive than of others. Many efforts have aimed at understanding the morphogenetic event of chondrogenesis in normal individuals, of which the main morphogenetic and regulatory mechanisms will be reviewed here. For instance, many signaling molecules that guide chondrogenesis--for example, transforming growth factor-beta, bone morphogenetic proteins, fibroblast growth factors, and Wnts, as well as transcriptional regulators such as the Sox family--have already been identified. Moreover, extracellular matrix components also play an important role in this developmental event, as evidenced by the promotion of the chondrogenic potential of chondroprogenitor cells caused by collagen II and proteoglycans like versican. The growing evidence of the elements that control chondrogenesis and the increasing number of different sources of progenitor cells will, hopefully, help to create tissue engineering platforms that could overcome many developmental or degenerative diseases associated with cartilage defects.
Collapse
Affiliation(s)
- Lluís Quintana
- Tissue Engineering Division, Department of Bioengineering, IQS-Ramon Llull University, Barcelona, Spain
| | | | | |
Collapse
|
550
|
Kisiday JD, Lee JH, Siparsky PN, Frisbie DD, Flannery CR, Sandy JD, Grodzinsky AJ. Catabolic responses of chondrocyte-seeded peptide hydrogel to dynamic compression. Ann Biomed Eng 2009; 37:1368-75. [PMID: 19415495 DOI: 10.1007/s10439-009-9699-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 04/11/2009] [Indexed: 01/12/2023]
Abstract
This study investigated the role of matrix metalloproteases and aggrecanases during dynamic compression-induced aggrecan catabolism in chondrocyte-seeded self-assembling peptide hydrogel. One- to two-week-old bovine chondrocytes were encapsulated into peptide hydrogel and cultured for 14 days prior to the application of an alternate day loading protocol. Dynamic compression-induced aggrecan catabolism was explored by evaluating GAG loss to the culture medium, zymography for matrix metalloproteases (MMPs), gene expression of MMPs and ADAMTS proteases, and Western blot analysis for aggrecan fragments. The application of loading over 4 days increased GAG loss to the medium three- to four-fold relative to free-swelling controls. Zymogram analysis detected increased concentrations of latent MMP-9 and MMP-3 in the culture medium relative to free-swelling culture. Real-time PCR showed expression levels of MMPs and ADAMTS proteases in loaded samples that ranged from 2.5- to 95-fold higher than free-swelling culture. Aggrecan fragment analysis did not detect small (50-80 kDa) molecular weight fragments in free-swelling culture; however, dynamic compression samples contained 60-80 kDa fragments that were detected by both anti-G1 and NITEGE probes, demonstrating ADAMTS but not MMP degradation. These data suggest that partially mature cartilage tissue engineering constructs may be susceptible to catabolic degradation.
Collapse
Affiliation(s)
- John D Kisiday
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | | | | | | | | | | | | |
Collapse
|