151
|
Pulkkinen H, Tiitu V, Valonen P, Hämäläinen ER, Lammi M, Kiviranta I. Recombinant human type II collagen as a material for cartilage tissue engineering. Int J Artif Organs 2018; 31:960-9. [DOI: 10.1177/039139880803101106] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose Collagen type II is the major component of cartilage and would be an optimal scaffold material for reconstruction of injured cartilage tissue. In this study, the feasibility of recombinant human type II collagen gel as a 3-dimensional culture system for bovine chondrocytes was evaluated in vitro. Methods Bovine chondrocytes (4x106 cells) were seeded within collagen gels and cultivated for up to 4 weeks. The gels were investigated with confocal microscopy, histology, and biochemical assays. Results Confocal microscopy revealed that the cells maintained their viability during the entire cultivation period. The chondrocytes were evenly distributed inside the gels, and the number of cells and the amount of the extracellular matrix increased during cultivation. The chondrocytes maintained their round phenotype during the 4-week cultivation period. The glycosaminoglycan levels of the tissue increased during the experiment. The relative levels of aggrecan and type II collagen mRNA measured with realtime polymerase chain reaction (PCR) showed an increase at 1 week. Conclusion Our results imply that recombinant human type II collagen is a promising biomaterial for cartilage tissue engineering, allowing homogeneous distribution in the gel and biosynthesis of extracellular matrix components.
Collapse
Affiliation(s)
- H.J. Pulkkinen
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
| | - V. Tiitu
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
| | - P. Valonen
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
| | - E.-R. Hämäläinen
- Bioprocess Engineering Laboratory, University of Oulu, Oulu - Finland
| | - M.J. Lammi
- Institute of Biomedicine, Department of Anatomy, University of Kuopio, Kuopio - Finland
- Department of Biosciences, Applied Biotechnology, University of Kuopio, Kuopio - Finland
| | - I. Kiviranta
- Department of Orthopaedics and Traumatology, Jyväskylä Central Hospital, Jyväskylä - Finland
- Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki - Finland
| |
Collapse
|
152
|
Pugliese R, Fontana F, Marchini A, Gelain F. Branched peptides integrate into self-assembled nanostructures and enhance biomechanics of peptidic hydrogels. Acta Biomater 2018; 66:258-271. [PMID: 29128535 DOI: 10.1016/j.actbio.2017.11.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
Self-assembling peptides (SAP) have drawn an increasing interest in the tissue engineering community. They display unquestionable biomimetic properties, tailorability and promising biocompatibility. However their use has been hampered by poor mechanical properties making them fragile soft scaffolds. To increase SAP hydrogel stiffness we introduced a novel strategy based on multiple ramifications of (LDLK)3, a well-known linear SAP, connected with one or multiple "lysine knots". Differently branched SAPs were tested by increasing the number of (LDLK)3-like branches and by adding the neuro-regenerative functional motif BMHP1 as a single branch. While pure branched peptides did not have appealing self-assembling propensity, when mixed with the corresponding linear SAP they increased the stiffness of the overall hydrogel of multiple times. Notably, optimal results (or peak) were obtained 1) at similar molar ratio (between linear and branched peptides) for all tested sequences and 2) for the branched SAPs featuring the highest number of branches made of (LDLK)3. The functional motif BMHP1, as expected, seemed not to contribute to the increase of the storage modulus as efficiently as (LDLK)3. Interestingly, branched SAPs improved the β-sheet self-arrangement of (LDLK)3 and allowed for the formation of assembled nanofibers. Indeed in coarse-grained molecular dynamics we showed they readily integrate in the assembled aggregates providing "molecular connections" among otherwise weakly paired β-structures. Lastly, branched SAPs did not affect the usual response of human neural stem cells cultured on (LDLK)3-like scaffolds in vitro. Hence, branched SAPs may be a valuable new tool to enhance mechanical properties of self-assembling peptide biomaterials harmlessly; as neither chemical nor enzymatic cross-linking reactions are involved. As a consequence, branched SAPs may enlarge the field of application of SAPs in tissue engineering and beyond. STATEMENT OF SIGNIFICANCE Self-assembling peptides stand at the forefront of regenerative medicine because they feature biomimetic nano-architectures that mimic the complexity of natural peptide-based extracellular matrices of living tissues. Their superior biocompatibility and ease of scale-up production are hampered by weak mechanical properties due to transient non-covalent interactions among and within the self-assembled peptide chains, thus limiting their potential applications. We introduced new branched self-assembling peptides to be used as "molecular connectors" among self-assembled nanostructures made of linear SAPs. Branched SAPs could be mixed with linear SAPs before self-assembling in order to have them intermingled with different β-sheets of linear SAPs after gelation. This strategy caused a manifold increase of the stiffness of the assembled hydrogels (proportional to the number of self-assembling branches), did not affect SAP propensity to form β-sheet but, instead, further stimulated their secondary structure rearrangements. It is now possible to modularly improve SAP scaffold mechanical properties without using harmful chemical reactions. Therefore, branched SAPs represent an additional tool to be adopted for efficient and harmless SAP scaffold customization in tissue engineering.
Collapse
|
153
|
Horkay F, Basser PJ, Hecht AM, Geissler E. Microstructure and Dynamic Properties of Aggrecan Assemblies. ACTA ACUST UNITED AC 2018. [DOI: 10.1557/adv.2018.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
154
|
Tsuang YH, Lin YS, Chen LT, Cheng CK, Sun JS. Effect of Dynamic Compression on in vitro Chondrocyte Metabolism. Int J Artif Organs 2018; 31:439-49. [DOI: 10.1177/039139880803100510] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Background Chondrocytes can detect and respond to the mechanical environment by altering their metabolism. This study was designed to explore the effects of dynamic compression on chondrocyte metabolism. Methods Chondrocytes were harvested from newborn Wistar rats. After 7 days of expansion, chondrocytes embedded in agarose discs underwent uniaxial unconfined dynamic compression loads at different amplitudes (5%, 10%, and 15%) and frequencies (0.5 Hz, 1.0 Hz, 2.0 Hz, and 3.0 Hz) with a duration of 24 hours. The delayed effects on the chondrocytes were studied at 1, 3, and 7 days after the experiment. Results The results showed that at 10% strain, higher-frequency compression pressure can enhance the proliferation of chondrocytes. The synthesis of glycosaminoglycan (GAG) increased at 10%-15% strain and a 1-Hz load. The synthesis of nitric oxide (NO) increased at the 0.5-Hz load; while decreasing at the 15% strain. With 10% strain, 1 Hz dynamic compression, the proliferation of chondrocytes and GAG synthesis increased and persisted for 7 days, and NO synthesis decreased at the third and seventh days of culture. Conclusions This study showed that chondrocytes respond metabolically to compressive loading, which is expected to modulate the growth and the resultant biomechanical properties of these tissue-engineered constructs during culture.
Collapse
Affiliation(s)
- Y.-H. Tsuang
- Department of Orthopedic Surgery, Taipei City Hospital, Taipei, Taiwan - ROC
| | - Y.-S. Lin
- Institute of Biomedical Engineering, National Yang-Ming University, Taipei, Taiwan - ROC
| | - L.-T. Chen
- Department of Research and Development, Healthbanks Biotechnology Corporation, Taipei, Taiwan - ROC
| | - C.-K. Cheng
- Institute of Biomedical Engineering, National Yang-Ming University, Taipei, Taiwan - ROC
| | - J.-S. Sun
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan - ROC
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan - ROC
| |
Collapse
|
155
|
Ando K, Imagama S, Kobayashi K, Ito K, Tsushima M, Morozumi M, Tanaka S, Machino M, Ota K, Nishida K, Nishida Y, Ishiguro N. Effects of a self-assembling peptide as a scaffold on bone formation in a defect. PLoS One 2018; 13:e0190833. [PMID: 29304115 PMCID: PMC5755907 DOI: 10.1371/journal.pone.0190833] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 12/20/2017] [Indexed: 11/19/2022] Open
Abstract
Spinal fusion and bone defect after injuries, removal of bone tumors, and infections need to be repaired by implantation. In an aging society, recovery from these procedures is often difficult. In this study, we found that injection of SPG-178 leads to expression of several bone marker genes and mineralization in vitro, and revealed a significantly higher degree of newly formed bone matrix with use of SPG-178 in vivo. MC3T3-E1 cells were used to evaluate osteoblast differentiation promoted by SPG-178. To analyze gene expression, total RNA was isolated from MC3T3-E1 cells cultured for 7 and 14 days with control medium or SPG-178 medium. Among the several bone marker genes examined, SPG-178 significantly increased the mRNA levels for ALP, BMP-2 and Osteocalcin, OPN, BSP and for the Osterix. Ten-week-old female Wistar rats were used for all transplantation procedures. A PEEK cage was implanted into a bony defect (5 mm) within the left femoral mid-shaft, and stability was maintained by an external fixator. The PEEK cages were filled with either a SPG-178 hydrogel plus allogeneic bone chips (n = 4) or only allogeneic bone chips (n = 4). The rats were then kept for 56 days. Newly formed bone matrix was revealed inside the PEEK cage and there was an increased bone volume per total volume with the cage filled with SPG-178, compared to the control group. SPG-178 has potential in clinical applications because it has several benefits. These include its favorable bone conduction properties its ability to act as a support for various different cells and growth factors, its lack of infection risk compared with materials of animal origin such as ECM, and the ease with which it can be used to fill defects with complex shapes and combined with a wide range of other materials.
Collapse
Affiliation(s)
- Kei Ando
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Shiro Imagama
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
- * E-mail:
| | - Kazuyoshi Kobayashi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Kenyu Ito
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Mikito Tsushima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Masayoshi Morozumi
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Satoshi Tanaka
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Masaaki Machino
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Kyotaro Ota
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Koji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshihiro Nishida
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| | - Naoki Ishiguro
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Aichi, Japan
| |
Collapse
|
156
|
Sever M, Tansik G, Arslan E, Yergoz F, Ozkan AD, Tekinay AB, Guler MO. Self-assembled peptide nanostructures and their gels for regenerative medicine applications. SELF-ASSEMBLING BIOMATERIALS 2018:455-473. [DOI: 10.1016/b978-0-08-102015-9.00022-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
157
|
Ni Y, Tang Z, Yang J, Gao Y, Lin H, Guo L, Zhang K, Zhang X. Collagen structure regulates MSCs behavior by MMPs involved cell–matrix interactions. J Mater Chem B 2018; 6:312-326. [DOI: 10.1039/c7tb02377d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Various scaffolds have been studied in the formation of cell niches and regulation of mesenchymal stem cells (MSCs) behaviors.
Collapse
Affiliation(s)
- Yilu Ni
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Zhurong Tang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Jirong Yang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yongli Gao
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Hai Lin
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Likun Guo
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
158
|
Das P, Pan I, Cohen E, Reches M. Self-assembly of a metallo-peptide into a drug delivery system using a “switch on” displacement strategy. J Mater Chem B 2018; 6:8228-8237. [DOI: 10.1039/c8tb01483c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Two newly designed tripeptides and their corresponding Cu2+ conjugates self-assemble into nanometric structures of different morphologies. These self-assembled metallo-peptide networks can serve as a drug delivery platform using a fluorescent-based "Turn-On" displacement strategy.
Collapse
Affiliation(s)
- Priyadip Das
- Institute of Chemistry and The Center for Nanoscience and Nanotechnology
- The Hebrew University of Jerusalem
- Jerusalem
- Israel
- SRM Research Institute
| | - Ieshita Pan
- Biochemistry and Molecular Biology
- Institute for Medical Research Israel-Canada
- The Hebrew University of Jerusalem
- Jerusalem
- Israel
| | - Ehud Cohen
- Biochemistry and Molecular Biology
- Institute for Medical Research Israel-Canada
- The Hebrew University of Jerusalem
- Jerusalem
- Israel
| | - Meital Reches
- Institute of Chemistry and The Center for Nanoscience and Nanotechnology
- The Hebrew University of Jerusalem
- Jerusalem
- Israel
| |
Collapse
|
159
|
Fontana G, Delgado LM, Cigognini D. Biologically Inspired Materials in Tissue Engineering. EXTRACELLULAR MATRIX FOR TISSUE ENGINEERING AND BIOMATERIALS 2018. [DOI: 10.1007/978-3-319-77023-9_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
160
|
Biomimetic Self-Assembling Peptide Hydrogels for Tissue Engineering Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:297-312. [DOI: 10.1007/978-981-13-0445-3_18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
161
|
Arad E, Malishev R, Rapaport H, Jelinek R. Membrane Determinants Affect Fibrillation Processes of β-Sheet Charged Peptides. Biomacromolecules 2017; 19:307-314. [DOI: 10.1021/acs.biomac.7b01318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Elad Arad
- Department of Chemistry, ‡Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, and §Ilse Katz Institute for Nano-Science and Technology (IKI), Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ravit Malishev
- Department of Chemistry, ‡Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, and §Ilse Katz Institute for Nano-Science and Technology (IKI), Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Hanna Rapaport
- Department of Chemistry, ‡Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, and §Ilse Katz Institute for Nano-Science and Technology (IKI), Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Raz Jelinek
- Department of Chemistry, ‡Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, and §Ilse Katz Institute for Nano-Science and Technology (IKI), Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
162
|
Taghavi L, Aramvash A, Seyedkarimi MS, Malek Sabet N. Evaluation of the hemocompatibility of RADA 16-I peptide. J Biomater Appl 2017; 32:1024-1031. [PMID: 29249197 DOI: 10.1177/0885328217748861] [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] [Indexed: 11/15/2022]
Abstract
RADA 16-I is an ionic self-assembling peptide that can form macroscopic scaffolds through β-sheet structures which are used in favor of cell growth and tissue engineering. This peptide has also the ability to stop bleeding effectively and quickly (∼20 seconds) when applied directly to the injuries. This study is focused on coagulation process, platelet aggregation, C3 and C4 concentrations, CBC counting, hemolysis, and white blood cell morphology tests to analyze hemocompatibility of RADA 16-I at different concentrations - 0.1, 0.2, 0.3 and 0.5%. According to the results, RADA 16-I hydrogel decreased the number of blood cells, slightly increased clot formation time and platelet aggregation, and yielded negligible hemolysis and only small changes in C3 and C4 concentrations and white blood cell morphology. All by all, the in vitro tests of hemocompatibility showed no perturbation in the blood composition when the peptides were in contact with the blood. The observed rapid hemostasis might be a result of increasing local concentrations of molecules involved in the formation of clot near the peptide hydrogel, thereby making a barrier which ended up with complete hemostasis. In conclusion, our experiments strongly supported further development of biomaterials based on RADA 16-I peptide.
Collapse
Affiliation(s)
- Laleh Taghavi
- Department of Bioscience and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| | - Asieh Aramvash
- Department of Bioscience and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| | | | - Narges Malek Sabet
- Department of Bioscience and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| |
Collapse
|
163
|
Abstract
Transcatheter arterial or venous embolization has been widely used to address solid tumors by occluding the tumor-feeding vessels. It is also performed to treat portosystemic shunts and to stop bleeding by repair of the site of trauma. Commonly used embolic materials are gelatin sponges, coils, beads, and liquid agents such as absolute ethanol, histoacyryl, and onyx. In the field of interventional radiology, embolotherapy is performed routinely. Liquid embolization agents have different characteristics. Their coagulation time, the inflammatory reaction of the vascular wall or surrounding tissue, and their adhesion to the vascular wall vary. PuraMatrix, a liquid embolic agent not yet available for clinical use, is comprised of amino acid. We introduce and discuss preliminary experimental studies to examine its potential for use in humans.
Collapse
Affiliation(s)
- Yasutaka Baba
- a Department of Diagnostic Radiology , Hiroshima University , Hiroshima , Japan
| | - Michiyo Higashi
- b Department of Human Pathology , Kagoshima University , Kagoshima , Japan
| | - Kazuo Awai
- a Department of Diagnostic Radiology , Hiroshima University , Hiroshima , Japan
| |
Collapse
|
164
|
Abstract
Angiogenesis plays an important role not only in the growth and regeneration of tissues in humans but also in pathological conditions such as inflammation, degenerative disease and the formation of tumors. Angiogenesis is also vital in thick engineered tissues and constructs, such as those for the heart and bone, as these can face difficulties in successful implantation if they are insufficiently vascularized or unable to connect to the host vasculature. Considerable research has been carried out on angiogenic processes using a variety of approaches. Pathological angiogenesis has been analyzed at the cellular level through investigation of cell migration and interactions, modeling tissue level interactions between engineered blood vessels and whole organs, and elucidating signaling pathways involved in different angiogenic stimuli. Approaches to regenerative angiogenesis in ischemic tissues or wound repair focus on the vascularization of tissues, which can be broadly classified into two categories: scaffolds to direct and facilitate tissue growth and targeted delivery of genes, cells, growth factors or drugs that promote the regeneration. With technological advancement, models have been designed and fabricated to recapitulate the innate microenvironment. Moreover, engineered constructs provide not only a scaffold for tissue ingrowth but a reservoir of agents that can be controllably released for therapeutic purposes. This review summarizes the current approaches for modeling pathological and regenerative angiogenesis in the context of micro-/nanotechnology and seeks to bridge these two seemingly distant aspects of angiogenesis. The ultimate aim is to provide insights and advances from various models in the realm of angiogenesis studies that can be applied to clinical situations.
Collapse
Affiliation(s)
- Li-Jiun Chen
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan.
| | | |
Collapse
|
165
|
Burgess KA, Miller AF, Oceandy D, Saiani A. Western blot analysis of cells encapsulated in self-assembling peptide hydrogels. Biotechniques 2017; 63:253-260. [DOI: 10.2144/000114617] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/19/2017] [Indexed: 11/23/2022] Open
Abstract
Continuous optimization of in vitro analytical techniques is ever more important, especially given the development of new materials for tissue engineering studies. In particular, isolation of cellular components for downstream applications is often hindered by the presence of biomaterials, presenting a major obstacle in understanding how cell–matrix interactions influence cell behavior. Here, we describe an approach for western blot analysis of cells that have been encapsulated in self-assembling peptide hydrogels (SAPHs), which highlights the need for complete solubilization of the hydrogel construct. We demonstrate that both the choice of buffer and multiple cycles of sonication are vital in obtaining complete solubilization, thereby enabling the detection of proteins otherwise lost to SAP aggregation. Moreover, we show that the presence of self-assembling peptides (SAPs) does not interfere with the standard immunoblotting technique, offering the potential for use in more full-scale proteomic studies.
Collapse
Affiliation(s)
- Kyle A. Burgess
- School of Materials, The University of Manchester, Manchester, UK
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Aline F. Miller
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
- School of Chemical Engineering and Analytical Sciences, The University of Manchester, Manchester, UK
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | - Alberto Saiani
- School of Materials, The University of Manchester, Manchester, UK
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| |
Collapse
|
166
|
Abstract
Bioinks, 3D cell culture systems which can be printed, are still in the early development stages. Currently, extensive research is going into designing printers to be more accommodating to bioinks, designing scaffolds with stiff materials as support structures for the often soft bioinks, and modifying the bioinks themselves. Recombinant spider silk proteins, a potential biomaterial component for bioinks, have high biocompatibility, can be processed into several morphologies and can be modified with cell adhesion motifs to enhance their bioactivity. In this work, thermally gelled hydrogels made from recombinant spider silk protein encapsulating mouse fibroblast cell line BALB/3T3 were prepared and characterized. The bioinks were evaluated for performance in vitro both before and after printing, and it was observed that unprinted bioinks provided a good platform for cell spreading and proliferation, while proliferation in printed scaffolds was prohibited. To improve the properties of the printed hydrogels, gelatin was given as an additive and thereby served indirectly as a plasticizer, improving the resolution of printed strands. Taken together, recombinant spider silk proteins and hydrogels made thereof show good potential as a bioink, warranting further development.
Collapse
Affiliation(s)
- Elise DeSimone
- Lehrstuhl Biomaterialien, Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayreuther Zentrum für Bio-Makromoleküle (bio-mac), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Bayerisches Polymerinstitut (BPI) Universitätsstraße 30, Universität Bayreuth, Bayreuth D-95447, Germany
| | | | | | | |
Collapse
|
167
|
Rothdiener M, Uynuk-Ool T, Südkamp N, Aurich M, Grodzinsky AJ, Kurz B, Rolauffs B. Human osteoarthritic chondrons outnumber patient- and joint-matched chondrocytes in hydrogel culture-Future application in autologous cell-based OA cartilage repair? J Tissue Eng Regen Med 2017; 12:e1206-e1220. [PMID: 28714570 DOI: 10.1002/term.2516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 06/09/2017] [Accepted: 07/11/2017] [Indexed: 12/22/2022]
Abstract
Autologous chondrocyte implantation (ACI) is used in 34-60% for osteoarthritic (OA) cartilage defects, although ACI is neither recommended nor designed for OA. Envisioning a hydrogel-based ACI for OA that uses chondrons instead of classically used chondrocytes, we hypothesized that human OA chondrons may outperform OA chondrocytes. We compared patient- and joint surface-matched human OA chondrons with OA chondrocytes cultured for the first time in a hydrogel, using a self-assembling peptide system. We determined yield, viability, cell numbers, mRNA expression, GAPDH mRNA enzyme activity, Collagen II synthesis (CPII) and degradation (C2C), and sulfated glycosaminoglycan. Ex vivo, mRNA expression was comparable. Over time, significant differences in survival led to 3.4-fold higher OA chondron numbers in hydrogels after 2 weeks (p = .002). Significantly, more enzymatically active GAPDH protein indicated higher metabolic activity. The number of cultures that expressed mRNA for Collagen Types I and VI, COMP, aggrecan, VEGF, TGF-β1, and FGF-2 (but not Collagen Types II and X) was different, resulting in a 3.5-fold higher number of expression-positive OA chondron cultures (p < .05). Measuring CPII and C2C per hydrogel, OA chondron hydrogels synthesized more than they degraded Collagen Type II, the opposite was true for OA chondrocytes. Per cell, OA chondrons but not OA chondrocytes displayed more synthesis than degradation. Thus, OA chondrons displayed superior biosynthesis and mRNA expression of tissue engineering and phenotype-relevant genes. Moreover, human OA chondrons displayed a significant survival advantage in hydrogel culture, whose presence, drastic extent, and timescale was novel and is clinically significant. Collectively, these data highlight the high potential of human OA chondrons for OA ACI, as they would outnumber and, thus, surpass OA chondrocytes.
Collapse
Affiliation(s)
- Miriam Rothdiener
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic, Eberhard Karls University, Tuebingen, Germany
| | - Tatiana Uynuk-Ool
- Siegfried Weller Institute for Trauma Research, BG Trauma Clinic, Eberhard Karls University, Tuebingen, Germany
| | - Norbert Südkamp
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, University Medical Center Freiburg, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Matthias Aurich
- Department of Orthopaedic and Trauma Surgery, Sana Kliniken Leipziger Land, Borna, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Universitätsklinikum Jena, Jena, Germany.,Department of Biochemistry, Rush Medical College, Chicago, IL, USA
| | - Alan J Grodzinsky
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bodo Kurz
- Department of Anatomy, Christian Albrechts University, Kiel, Germany
| | - Bernd Rolauffs
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, University Medical Center Freiburg, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
168
|
Abstract
As a novel class of biomaterials, nucleopeptides, via the conjugation of nucleobases and peptides, usually self-assemble to form nanofibres driven mainly by hydrogen bonds. Containing nucleobase(s), nucleopeptides have a unique property-interacting with nucleic acids. Here we report the design and characterization of nucleopeptides that self-assemble in water and are able to interact with single-stranded DNAs (ssDNAs). Containing nucleobases on their side chains, these nucleopeptides bind with the ssDNAs, and the ssDNAs reciprocally affect the self-assembly of nucleopeptides. In addition, the interactions between nucleopeptides and ssDNAs also decrease their proteolytic resistance against proteinase K, which further demonstrates the binding with ssDNAs. The nucleopeptides also interact with plasmid DNA and deliver hairpin DNA into cells. This work illustrates a new and rational approach to create soft biomaterials by the integration of nucleobases and peptides to bind with DNA, which may lead to the development of nucleopeptides for controlling DNA in cells.
Collapse
Affiliation(s)
- Xuewen Du
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Jie Zhou
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Xinming Li
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| |
Collapse
|
169
|
Abstract
Peptides are ubiquitous in nature and useful in many fields, from agriculture as pesticides, in medicine as antibacterial and antifungal drugs founded in the innate immune systems, to medicinal chemistry as hormones. However, the concept of peptides as materials was not recognized until 1990 when a self-assembling peptide as a repeating segment in a yeast protein was serendipitously discovered. Peptide materials are so called because they have bona fide materials property and are made from simple amino acids with well-ordered nanostructures under physiological conditions. These structures include well-ordered nanofibres, nanotubes and nanovesicles. These peptide materials have been used for: (i) three-dimensional tissue cell cultures of primary cells and stem cells, (ii) three-dimensional tissue printing, (iii) sustained releases of small molecules, growth factors, monoclonal antibody and siRNA, (iv) accelerated wound healing in reparative and regenerative medicine as well as tissue engineering, (v) used to stabilize membrane proteins including difficult G-protein coupled receptors and photosystem I for designing nanobiodevices, (vi) a few self-assembling peptides have been used in human clinical trials for accelerated wound healings in surgical uses and (vii) in human clinical trials for siRNA delivery for treatment of cancers. It is likely that these self-assembling peptides will open doors for more and more diverse uses. The field of self-assembling peptides is growing in a number of directions in areas of materials, synthetic biology, and clinical medicine and beyond.
Collapse
Affiliation(s)
- Shuguang Zhang
- Laboratory of Molecular Architecture, Canter for Bits and Atoms, Massachusetts Institute of Technology, Cambridge, MA 02139-4307, USA
| |
Collapse
|
170
|
Development of Multilayered Chlorogenate-Peptide Based Biocomposite Scaffolds for Potential Applications in Ligament Tissue Engineering - An <i>In Vitro</i> Study. JOURNAL OF BIOMIMETICS BIOMATERIALS AND BIOMEDICAL ENGINEERING 2017. [DOI: 10.4028/www.scientific.net/jbbbe.34.37] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this work, for the first time, chlorogenic acid, a natural phytochemical, was conjugated to a lactoferrin derived antimicrobial peptide sequence RRWQWRMKKLG to develop a self-assembled template. To mimic the components of extracellular matrix, we then incorporated Type I Collagen, followed by a sequence of aggrecan peptide (ATEGQVRVNSIYQDKVSL) onto the self-assembled templates for potential applications in ligament tissue regeneration. Mechanical properties and surface roughness were studied and the scaffolds displayed a Young’s Modulus of 169 MP and an average roughness of 72 nm respectively. Thermal phase changes were studied by DSC analysis. Results showed short endothermic peaks due to water loss and an exothermic peak due to crystallization of the scaffold caused by rearrangement of the components. Biodegradability studies indicated a percent weight loss of 27.5 % over a period of 37 days. Furthermore, the scaffolds were found to adhere to fibroblasts, the main cellular component of ligament tissue. The scaffolds promoted cell proliferation and displayed actin stress fibers indicative of cell motility and attachment. Collagen and proteoglycan synthesis were also promoted, demonstrating increased expression and deposition of collagen and proteoglycans. Additionally, the scaffolds exhibited antimicrobial activity against Staphylococcus epidermis bacteria, which is beneficial for minimizing biofilm formation if potentially used as implants. Thus, we have developed a novel biocomposite that may open new avenues to enhance ligament tissue regeneration.
Collapse
|
171
|
Chatterjee S, Maitra U. In situ formation of luminescent CdSe QDs in a metallohydrogel: a strategy towards synthesis, isolation, storage and re-dispersion of the QDs. NANOSCALE 2017; 9:13820-13827. [PMID: 28891578 DOI: 10.1039/c7nr03758a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A one step, in situ, room temperature synthesis of yellow luminescent CdSe QDs was achieved in a metallohydrogel derived from a facially amphiphilic bile salt, resulting in a QD-gel hybrid. An ordered self-assembly and homogeneous distribution of the CdSe QDs in the hydrogel network was observed from optical and electron micrographs. The different excited state behavior of the CdSe QDs in the hybrid was revealed for the first time using time resolved spectroscopy. We also describe the successful isolation of the photoluminescent CdSe QDs from the gel followed by their re-dispersion in an organic solvent using suitable capping ligands.
Collapse
Affiliation(s)
- Sayantan Chatterjee
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012 Karnataka, India.
| | | |
Collapse
|
172
|
Singh R, Bellat V, Wang M, Schweitzer ME, Wu YL, Tung CH, Souweidane MM, Law B. Volume of distribution and clearance of peptide-based nanofiber after convection-enhanced delivery. J Neurosurg 2017; 129:10-18. [PMID: 28885119 DOI: 10.3171/2017.2.jns162273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Drug clearance may be a limiting factor in the clinical application of convection-enhanced delivery (CED). Peptide-based nanofibers (NFPs) have a high aspect ratio, and NFPs loaded with drugs could potentially maintain effective drug concentrations for an extended period sufficient for cancer therapy. The objective of this study was to assess the volume of distribution (Vd) and clearance of variable lengths of NFPs when administered using CED. METHODS NFPs composed of multiple methoxypolyethylene glycol (mPEG)-conjugated constructs (mPEG2000-KLDLKLDLKLDL-K( FITC)-CONH2, for which FITC is fluorescein isothiocyanate) were assembled in an aqueous buffer. The NFPs were approximately 5 nm in width and were formulated into different lengths: 100 nm (NFP-100), 400 nm (NFP-400), and 1000 nm (NFP-1000). The NFP surface was covalently conjugated with multiple Cy5.5 fluorophores as the optical reporters to track the post-CED distribution. Forty-two 6- to 8-week-old Ntv-a;p53fl/fl mice underwent CED to the striatum. Animals were killed immediately, 24 hours or 72 hours after CED. The brains were extracted and sectioned for assessing NFP Vd to volume of infusion (Vi) ratio, and clearance using fluorescence microscopy. RESULTS CED of NFPs was well tolerated by all the animals. The average Vd/Vi ratios for NFP-100, NFP-400, NFP-1000, and unconjugated positive control (free Cy5.5) were 1.87, 2.47, 1.07, and 3.0, respectively, which were statistically different (p = 0.003). The percentages remaining of the original infusion volume at 24 hours for NFP-100, -400, and -1000 were 40%, 90%, and 74%, respectively. The percentages remaining at 72 hours for NFP-100, -400, and -1000 were 15%, 30%, and 46%, respectively. Unconjugated Cy5.5 was not detected at 24 or 72 hours after CED. CONCLUSIONS CED of NFPs is feasible with Vd/Vi ratios and clearance rates comparable to other nanocarriers. Of the 3 NFPs, NFP-400 appears to provide the best distribution and slowest clearance after 24 hours. NFP provides a dynamic theranostic platform, with the potential to deliver clinically efficacious drug payload to brain tumor after CED.
Collapse
Affiliation(s)
| | - Vanessa Bellat
- 2Department of Radiology, Molecular Imaging Innovations Institute, and
| | | | | | | | - Ching-Hsuan Tung
- 2Department of Radiology, Molecular Imaging Innovations Institute, and
| | - Mark M Souweidane
- 1Department of Neurological Surgery.,3Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - Benedict Law
- 2Department of Radiology, Molecular Imaging Innovations Institute, and
| |
Collapse
|
173
|
Das S, Kumar R, Jha NN, Maji SK. Controlled Exposure of Bioactive Growth Factor in 3D Amyloid Hydrogel for Stem Cells Differentiation. Adv Healthc Mater 2017; 6. [PMID: 28736995 DOI: 10.1002/adhm.201700368] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/09/2017] [Indexed: 12/21/2022]
Abstract
Amyloid based hydrogels can mimic the extracellular matrix and serve as matrices for tissue engineering both in vitro and in vivo. A pH responsive self-assembled amyloid hydrogel system is used to encapsulate various growth factors for driving stem cell differentiation toward neuronal lineage. Diffusion studies with fluorescence recovery after photobleaching and bulk release with the model protein fluorescein isothiocyanate-bovine serum albumin show that encapsulated protein molecules can be released in a sustained fashion from the hydrogel over a considerable period of time (30 d). Moreover, by modulating the porosity of the hydrogel by the simple addition of salt, the encapsulated protein molecules can be retained for a longer period of time within the hydrogel. Mesenchymal stem cells, when cultured in 3D amyloid hydrogels with growth factors fibroblast growth factor 8 and sonic hedgehog, show more neuron specific differentiation as compared to hydrogel alone. This higher differentiation potential of growth factor encapsulated amyloid hydrogels can be due to concomitant exposure of cells to biomechanical as well as biochemical cues during the course of differentiation. The present study suggests that amyloid based hydrogel can be exploited for controlled growth factor delivery as well as directed stem cell differentiation to neuron.
Collapse
Affiliation(s)
- Subhadeep Das
- IITB‐Monash Research Academy Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
- Department of Materials Science and Engineering Monash University Clayton VIC 3800 Australia
| | - Rakesh Kumar
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
| | - Narendra Nath Jha
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering Indian Institute of Technology Bombay Mumbai Maharashtra 400076 India
| |
Collapse
|
174
|
Chen S, Zhou A, He B, Zhao W, Chen X, Jiang D. Designer D-form self-assembling peptide scaffolds promote the proliferation and migration of rat bone marrow-derived mesenchymal stem cells. Int J Mol Med 2017; 40:679-688. [PMID: 28677805 PMCID: PMC5547947 DOI: 10.3892/ijmm.2017.3056] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/23/2017] [Indexed: 01/06/2023] Open
Abstract
Self-assembling peptide (SAP) nanofiber hydrogel scaffolds have become increasingly important in tissue engineering due to their outstanding bioactivity and biodegradability. However, there is an initial concern on their long-term clinical use, since SAPs made of L-form amino acid sequences are sensitive to enzymatic degradation. In this study, we present a designer SAP, D-RADA16, made of all D-amino acid. We investigated the nanofiber morphology of D-RADA16, its potential for the culture of bone marrow-derived mesenchymal stem cells (BMSCs), and the proteolytic resistance of the biomaterial. The results revealed that D-RADA16 exhibited stable β-sheets and formed interwoven nanofiber scaffolds in water. D-RADA16 and L-RADA16 hydrogel scaffolds were both found to promote the proliferation and migration of rat BMSCs in the 3D cell culture microenvironment. Furthermore, the D-RADA16 scaffolds exhibited a higher proteolytic resistance against proteinase K than the L-RADA16 scaffolds. These observations indicate that D-RADA16 hydrogel scaffolds have excellent bioactivity, biocompatibility and biostability, and thus may serve as promising candidates for long-term application in vivo.
Collapse
Affiliation(s)
- Shuo Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Bin He
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaojun Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
175
|
Xia Y, Xue B, Qin M, Cao Y, Li Y, Wang W. Printable Fluorescent Hydrogels Based on Self-Assembling Peptides. Sci Rep 2017; 7:9691. [PMID: 28852128 PMCID: PMC5574881 DOI: 10.1038/s41598-017-10162-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/02/2017] [Indexed: 11/18/2022] Open
Abstract
Fluorescent hydrogels (FH) have a variety of potential applications in the field of soft electronics. However, fabrication of mechanically stable and printable fluorescent hydrogels remains challenging. Here, we report a kind of fluorescent hydrogel based on the co-assembly of peptide motif and transition metal ions. The metal ions are captured in the hydrogel network at specific positions through covalently linked ligands on the peptide hydrogelators. This efficiently prevents the aggregation and self-quenching of organometallic chromophores. In addition, the formation of metal-ligand complexes introduces additional interactions to stabilize the hydrogel network, making the FH even more stable after the incorporation of metal ions. The FH is optically transparent but highly fluorescent. By using three different metal ions, the white light fluorescent supramolecular hydrogel has been achieved. As a proof-of-principle, we demonstrate the printability of the hydrogels to various patterns. We anticipate that with the improved fluorescent performance and stability, this kind of FH can find broad applications in extrusion-based 3D printing for the construction of soft electronics.
Collapse
Affiliation(s)
- Yifan Xia
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P.R. China
| | - Bin Xue
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P.R. China.
| | - Meng Qin
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P.R. China
| | - Yi Cao
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P.R. China
| | - Ying Li
- Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, Jiangsu Engineering Technology Research Centre of Environmental Cleaning Materials, Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Jiangsu Joint Laboratory of Atmospheric Pollution Control, Jiangsu School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, 219 Ningliu Road, Nanjing, Jiangsu, 210044, P.R. China.
| | - Wei Wang
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, 22 Hankou Road, Nanjing, Jiangsu, 210093, P.R. China.
| |
Collapse
|
176
|
Jin W, Penington CJ, McCue SW, Simpson MJ. A computational modelling framework to quantify the effects of passaging cell lines. PLoS One 2017; 12:e0181941. [PMID: 28750084 PMCID: PMC5531485 DOI: 10.1371/journal.pone.0181941] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/10/2017] [Indexed: 11/28/2022] Open
Abstract
In vitro cell culture is routinely used to grow and supply a sufficiently large number of cells for various types of cell biology experiments. Previous experimental studies report that cell characteristics evolve as the passage number increases, and various cell lines can behave differently at high passage numbers. To provide insight into the putative mechanisms that might give rise to these differences, we perform in silico experiments using a random walk model to mimic the in vitro cell culture process. Our results show that it is possible for the average proliferation rate to either increase or decrease as the passaging process takes place, and this is due to a competition between the initial heterogeneity and the degree to which passaging damages the cells. We also simulate a suite of scratch assays with cells from near–homogeneous and heterogeneous cell lines, at both high and low passage numbers. Although it is common in the literature to report experimental results without disclosing the passage number, our results show that we obtain significantly different closure rates when performing in silico scratch assays using cells with different passage numbers. Therefore, we suggest that the passage number should always be reported to ensure that the experiment is as reproducible as possible. Furthermore, our modelling also suggests some avenues for further experimental examination that could be used to validate or refine our simulation results.
Collapse
Affiliation(s)
- Wang Jin
- School of Mathematical Sciences, Queensland University of Technology (QUT) Brisbane, Queensland 4000, Australia
| | - Catherine J. Penington
- School of Mathematical Sciences, Queensland University of Technology (QUT) Brisbane, Queensland 4000, Australia
| | - Scott W. McCue
- School of Mathematical Sciences, Queensland University of Technology (QUT) Brisbane, Queensland 4000, Australia
| | - Matthew J. Simpson
- School of Mathematical Sciences, Queensland University of Technology (QUT) Brisbane, Queensland 4000, Australia
- * E-mail:
| |
Collapse
|
177
|
Szloszár A, Fülöp F, Mándity IM. Accelerated Synthesis of Protected Peptides in a Continuous-Flow Fixed-Bed Reactor. ChemistrySelect 2017. [DOI: 10.1002/slct.201701489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Aliz Szloszár
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös u. 6 H-6720 Szeged Hungary
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös u. 6 H-6720 Szeged Hungary
- Stereochemistry Research Group; Hungarian Academy of Sciences; Eötvös u. 6 H-6720 Szeged Hungary
| | - István M. Mándity
- Institute of Pharmaceutical Chemistry; University of Szeged; Eötvös u. 6 H-6720 Szeged Hungary
| |
Collapse
|
178
|
Del Valle LJ, Díaz A, Puiggalí J. Hydrogels for Biomedical Applications: Cellulose, Chitosan, and Protein/Peptide Derivatives. Gels 2017; 3:E27. [PMID: 30920524 PMCID: PMC6318613 DOI: 10.3390/gels3030027] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/09/2017] [Accepted: 07/10/2017] [Indexed: 12/21/2022] Open
Abstract
Hydrogels based on polysaccharide and protein natural polymers are of great interest in biomedical applications and more specifically for tissue regeneration and drug delivery. Cellulose, chitosan (a chitin derivative), and collagen are probably the most important components since they are the most abundant natural polymers on earth (cellulose and chitin) and in the human body (collagen). Peptides also merit attention because their self-assembling properties mimic the proteins that are present in the extracellular matrix. The present review is mainly focused on explaining the recent advances on hydrogels derived from the indicated polymers or their combinations. Attention has also been paid to the development of hydrogels for innovative biomedical uses. Therefore, smart materials displaying stimuli responsiveness and having shape memory properties are considered. The use of micro- and nanogels for drug delivery applications is also discussed, as well as the high potential of protein-based hydrogels in the production of bioactive matrices with recognition ability (molecular imprinting). Finally, mention is also given to the development of 3D bioprinting technologies.
Collapse
Affiliation(s)
- Luís J Del Valle
- Barcelona Research Center for Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Escola d'Enginyeria de Barcelona Est-EEBE, c/Eduard Maristany 10-14, Barcelona 08019, Spain.
| | - Angélica Díaz
- Barcelona Research Center for Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Escola d'Enginyeria de Barcelona Est-EEBE, c/Eduard Maristany 10-14, Barcelona 08019, Spain.
| | - Jordi Puiggalí
- Barcelona Research Center for Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Escola d'Enginyeria de Barcelona Est-EEBE, c/Eduard Maristany 10-14, Barcelona 08019, Spain.
| |
Collapse
|
179
|
Mansour A, Mezour MA, Badran Z, Tamimi F. * Extracellular Matrices for Bone Regeneration: A Literature Review. Tissue Eng Part A 2017; 23:1436-1451. [PMID: 28562183 DOI: 10.1089/ten.tea.2017.0026] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The gold standard material for bone regeneration is still autologous bone, a mesenchymal tissue that consists mainly of extracellular matrix (ECM) (90% v/v) and little cellular content (10% v/v). However, the fact that decellularized allogenic bone grafts often present a clinical performance comparable to autologous bone grafts demonstrates the crucial role of ECM in bone regeneration. For long, the mechanism by which bone allografts function was not clear, but recent research has unveiled many unique characteristics of ECM that seem to play a key role in tissue regeneration. This is further confirmed by the fact that synthetic biomaterials with composition and properties resembling bone ECM present excellent bone regeneration properties. In this context, ECM molecules such as glycosaminoglycans (GAGs) and self-assembly peptides (SAPs) can improve the performance of bone regeneration biomaterials. Moreover, decellularized ECM derived either from native tissues such as bone, cartilage, skin, and tooth germs or from cells such as osteoblasts, chondrocytes, and stem cells has shown promising results in bone regeneration applications. Understanding the role of ECM in bone regeneration is crucial for the development of the next generation of biomaterials for bone tissue engineering. In this sense, this review addresses the state-of-the-art on this subject matter.
Collapse
Affiliation(s)
- Alaa Mansour
- 1 Faculty of Dentistry, McGill University , Montreal, Canada
| | | | - Zahi Badran
- 1 Faculty of Dentistry, McGill University , Montreal, Canada .,2 Department of Periodontology (CHU/UIC 11, INSERM UMR 1229-RMeS), Faculty of Dental Surgery, University of Nantes , Nantes, France
| | - Faleh Tamimi
- 1 Faculty of Dentistry, McGill University , Montreal, Canada
| |
Collapse
|
180
|
Peng S, Wu CW, Lin JY, Yang CY, Cheng MH, Chu IM. Promoting chondrocyte cell clustering through tuning of a poly(ethylene glycol)-poly(peptide) thermosensitive hydrogel with distinctive microarchitecture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:181-189. [DOI: 10.1016/j.msec.2017.02.130] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/05/2017] [Accepted: 02/24/2017] [Indexed: 01/14/2023]
|
181
|
Self-Assembling RADA16-I Peptide Hydrogel Scaffold Loaded with Tamoxifen for Breast Reconstruction. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3656193. [PMID: 28691024 PMCID: PMC5485292 DOI: 10.1155/2017/3656193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/11/2017] [Accepted: 05/03/2017] [Indexed: 12/20/2022]
Abstract
More and more breast cancer patients prefer autologous fat tissue transfer following lumpectomy to maintain perfect female characteristics. However, the outcome was not satisfactory due to the transplanted fat absorption. In this study, we prepared two RADA16-I peptide scaffolds with and without tamoxifen. Both scaffolds were transparent, porous, and hemisphere-shaped. The hADSCs isolated from liposuction were attached to the scaffold. The growth inhibition of the hADSCs induced by TAM in 2-demensional (2D) culture was higher than that in TAM-loaded hydrogel scaffold 3D culture (P < 0.05); however, the same outcomes were not observed in MCF-7 cells. Correspondingly, the apoptosis of the hADSCs induced by TAM was significantly increased in 2D culture compared to that in scaffold 3D culture (P < 0.05). Yet the outcomes of the aoptosis in MCF-7 were contrary. Apoptosis-related protein Bcl-2 was involved in the process. In vivo experiments showed that both scaffolds formed a round mass after subcutaneous implantation and it retained its shape after being pressed slightly. The implantation had no effect on the weight and activity of the animals. The results suggested that TAM-loaded RADA16-I hydrogel scaffolds both provide support for hADSCs cells attachment/proliferation and retain cytotoxic effect on MCF-7 cells, which might be a promising therapeutic breast tissue following lumpectomy.
Collapse
|
182
|
Controlled and sustained delivery of siRNA/NPs from hydrogels expedites bone fracture healing. Biomaterials 2017; 139:127-138. [PMID: 28601703 DOI: 10.1016/j.biomaterials.2017.06.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/16/2017] [Accepted: 06/02/2017] [Indexed: 01/01/2023]
Abstract
Despite great potential, delivery remains as the most significant barrier to the widespread use of siRNA therapeutics. siRNA has delivery limitations due to susceptibility to RNase degradation, low cellular uptake, and poor tissue-specific localization. Here, we report the development of a hybrid nanoparticle (NP)/hydrogel system that overcomes these challenges. Hydrogels provide localized and sustained delivery via controlled release of entrapped siRNA/NP complexes while NPs protect and enable efficient cytosolic accumulation of siRNA. To demonstrate therapeutic efficacy, regenerative siRNA against WW domain-containing E3 ubiquitin protein ligase 1 (Wwp1) complexed with NP were entrapped within poly(ethylene glycol) (PEG)-based hydrogels and implanted at sites of murine mid-diaphyseal femur fractures. Results showed localization of hydrogels and controlled release of siRNA/NPs at fractures for 28 days, a timeframe over which fracture healing occurs. siRNA/NP sustained delivery from hydrogels resulted in significant Wwp1 silencing at fracture callus compared to untreated controls. Fractures treated with siRNA/NP hydrogels exhibited accelerated bone formation and significantly increased biomechanical strength. This NP/hydrogel siRNA delivery system has outstanding therapeutic promise to augment fracture healing. Owing to the structural similarities of siRNA, the development of the hydrogel platform for in vivo siRNA delivery has myriad therapeutic possibilities in orthopaedics and beyond.
Collapse
|
183
|
Mc Donald CK, Moriarty P, Varzgalis M, Murphy C. The Top 50 Most Cited Articles in Cartilage Regeneration. Biores Open Access 2017; 6:58-62. [PMID: 28736688 PMCID: PMC5515090 DOI: 10.1089/biores.2017.0006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The aim of this study was to identify and analyze the top 50 most cited articles in cartilage regeneration. The impact of a scientific journal can be gauged by the total number of citations it has accrued. The top 50 most cited articles involving cartilage regeneration represent the most quoted level of evidence among this new subspecialty. This study aims to identify and analyze the 50 most cited articles in cartilage regeneration. The Web of Science™ citation indexing service was utilized to determine the most frequently cited articles published after 1956 containing “cartilage regeneration” in the “topic” or “title.” The 50 most cited articles were included. The number of citations, year of publication, country of article origin, article institution, journal of publication, publication format, and authorship were then calculated for each article. The span of citations ranged from 1287 to 203 citations, with a mean of 361.02 citations per article in question. The articles originated from 11 countries, with the United States contributing 34 articles, followed by Japan with 5 articles. The articles were distributed across 34 high-impact journals. Biomaterials was the journal with the highest number of publications (seven articles) followed by the Journal of Orthopaedic Research (three articles). Of the 50 articles, 2 were clinical observational studies, 47 concerned basic science, and 1 was review article. The most cited articles involving cartilage regeneration are detected in both experimental and clinical research fields. The high ratio of basic science to clinical articles reflects the infancy of this relatively new specialty and that further clinical research is required in this area.
Collapse
Affiliation(s)
- Ciaran K Mc Donald
- Department of Trauma and Orthopaedics, Galway University Hospital, Galway, Ireland
| | - Peter Moriarty
- Department of Trauma and Orthopaedics, Galway University Hospital, Galway, Ireland
| | - Manvydas Varzgalis
- Department of Breast & Endocrine Surgery, Galway University Hospital, Galway, Ireland
| | - Colin Murphy
- Department of Trauma and Orthopaedics, Galway University Hospital, Galway, Ireland
| |
Collapse
|
184
|
Camarero-Espinosa S, Cooper-White J. Tailoring biomaterial scaffolds for osteochondral repair. Int J Pharm 2017; 523:476-489. [DOI: 10.1016/j.ijpharm.2016.10.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
|
185
|
Guan X, Avci-Adali M, Alarçin E, Cheng H, Kashaf SS, Li Y, Chawla A, Jang HL, Khademhosseini A. Development of hydrogels for regenerative engineering. Biotechnol J 2017; 12:10.1002/biot.201600394. [PMID: 28220995 PMCID: PMC5503693 DOI: 10.1002/biot.201600394] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 11/07/2022]
Abstract
The aim of regenerative engineering is to restore complex tissues and biological systems through convergence in the fields of advanced biomaterials, stem cell science, and developmental biology. Hydrogels are one of the most attractive biomaterials for regenerative engineering, since they can be engineered into tissue mimetic 3D scaffolds to support cell growth due to their similarity to native extracellular matrix. Advanced nano- and micro-technologies have dramatically increased the ability to control properties and functionalities of hydrogel materials by facilitating biomimetic fabrication of more sophisticated compositions and architectures, thus extending our understanding of cell-matrix interactions at the nanoscale. With this perspective, this review discusses the most commonly used hydrogel materials and their fabrication strategies for regenerative engineering. We highlight the physical, chemical, and functional modulation of hydrogels to design and engineer biomimetic tissues based on recent achievements in nano- and micro-technologies. In addition, current hydrogel-based regenerative engineering strategies for treating multiple tissues, such as musculoskeletal, nervous and cardiac tissue, are also covered in this review. The interaction of multiple disciplines including materials science, cell biology, and chemistry, will further play an important role in the design of functional hydrogels for the regeneration of complex tissues.
Collapse
Affiliation(s)
- Xiaofei Guan
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Orthopedic Department, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstr. 7/1, Tuebingen 72076, Germany
| | - Emine Alarçin
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul 34668, Turkey
| | - Hao Cheng
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sara Saheb Kashaf
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuxiao Li
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aditya Chawla
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hae Lin Jang
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women’s Hospital, Boston, MA 02139, USA
- Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience & Technology, Konkuk University, Seoul 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
186
|
Pedde RD, Mirani B, Navaei A, Styan T, Wong S, Mehrali M, Thakur A, Mohtaram NK, Bayati A, Dolatshahi-Pirouz A, Nikkhah M, Willerth SM, Akbari M. Emerging Biofabrication Strategies for Engineering Complex Tissue Constructs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1606061. [PMID: 28370405 DOI: 10.1002/adma.201606061] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/16/2017] [Indexed: 05/24/2023]
Abstract
The demand for organ transplantation and repair, coupled with a shortage of available donors, poses an urgent clinical need for the development of innovative treatment strategies for long-term repair and regeneration of injured or diseased tissues and organs. Bioengineering organs, by growing patient-derived cells in biomaterial scaffolds in the presence of pertinent physicochemical signals, provides a promising solution to meet this demand. However, recapitulating the structural and cytoarchitectural complexities of native tissues in vitro remains a significant challenge to be addressed. Through tremendous efforts over the past decade, several innovative biofabrication strategies have been developed to overcome these challenges. This review highlights recent work on emerging three-dimensional bioprinting and textile techniques, compares the advantages and shortcomings of these approaches, outlines the use of common biomaterials and advanced hybrid scaffolds, and describes several design considerations including the structural, physical, biological, and economical parameters that are crucial for the fabrication of functional, complex, engineered tissues. Finally, the applications of these biofabrication strategies in neural, skin, connective, and muscle tissue engineering are explored.
Collapse
Affiliation(s)
- R Daniel Pedde
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Bahram Mirani
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Ali Navaei
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85281, USA
| | - Tara Styan
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Sarah Wong
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Mehdi Mehrali
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Kgs. Lyngby, 2800, Denmark
| | - Ashish Thakur
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Kgs. Lyngby, 2800, Denmark
| | - Nima Khadem Mohtaram
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Armin Bayati
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Alireza Dolatshahi-Pirouz
- Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, Technical University of Denmark, Kgs. Lyngby, 2800, Denmark
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85281, USA
| | - Stephanie M Willerth
- Willerth Laboratory, Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, V8P 5C2, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Center for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, V8P 5C2, Canada
- Center for Biomedical Research, University of Victoria, Victoria, V8P 5C2, Canada
| |
Collapse
|
187
|
Ansari S, Seagroves JT, Chen C, Shah K, Aghaloo T, Wu BM, Bencharit S, Moshaverinia A. Dental and orofacial mesenchymal stem cells in craniofacial regeneration: The prosthodontist's point of view. J Prosthet Dent 2017; 118:455-461. [PMID: 28385446 DOI: 10.1016/j.prosdent.2016.11.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022]
Abstract
Of the available regenerative treatment options, craniofacial tissue regeneration using mesenchymal stem cells (MSCs) shows promise. The ability of stem cells to produce multiple specialized cell types along with their extensive distribution in many adult tissues have made them an attractive target for applications in tissue engineering. MSCs reside in a wide spectrum of postnatal tissue types and have been successfully isolated from orofacial tissues. These dental- or orofacial-derived MSCs possess self-renewal and multilineage differentiation capacities. The craniofacial system is composed of complex hard and soft tissues derived from sophisticated processes starting with embryonic development. Because of the complexity of the craniofacial tissues, the application of stem cells presents challenges in terms of the size, shape, and form of the engineered structures, the specialized final developed cells, and the modulation of timely blood supply while limiting inflammatory and immunological responses. The cell delivery vehicle has an important role in the in vivo performance of stem cells and could dictate the success of the regenerative therapy. Among the available hydrogel biomaterials for cell encapsulation, alginate-based hydrogels have shown promising results in biomedical applications. Alginate scaffolds encapsulating MSCs can provide a suitable microenvironment for cell viability and differentiation for tissue regeneration applications. This review aims to summarize current applications of dental-derived stem cell therapy and highlight the use of alginate-based hydrogels for applications in craniofacial tissue engineering.
Collapse
Affiliation(s)
- Sahar Ansari
- Lecturer, Division of Oral Biology, School of Dentistry, University of California, Los Angeles, Calif
| | - Jackson T Seagroves
- Student, Department of Dental Research, School of Dentistry, University of North Carolina, Chapel Hill, NC
| | - Chider Chen
- Postdoctoral research fellow, Department of Anatomy and Cell Biology, School of Dental Medicine University of Pennsylvania, Philadelphia, Pa
| | - Kumar Shah
- Associate Professor and Program Director, Graduate Program in Prosthodontics, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Calif
| | - Tara Aghaloo
- Professor, Division of Advanced Prosthodontics and Director, Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, Calif
| | - Benjamin M Wu
- Professor and Chair, Division of Advanced Prosthodontics and Director, Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, Calif
| | - Sompop Bencharit
- Associate Professor and Director, Digital Dentistry Technologies, Department of General Practice and Department of Oral & Maxillofacial Surgery, School of Dentistry, and Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA
| | - Alireza Moshaverinia
- Assistant Professor, Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Calif.
| |
Collapse
|
188
|
Dou XQ, Feng CL. Amino Acids and Peptide-Based Supramolecular Hydrogels for Three-Dimensional Cell Culture. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1604062. [PMID: 28112836 DOI: 10.1002/adma.201604062] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Indexed: 05/18/2023]
Abstract
Supramolecular hydrogels assembled from amino acids and peptide-derived hydrogelators have shown great potential as biomimetic three-dimensional (3D) extracellular matrices because of their merits over conventional polymeric hydrogels, such as non-covalent or physical interactions, controllable self-assembly, and biocompatibility. These merits enable hydrogels to be made not only by using external stimuli, but also under physiological conditions by rationally designing gelator structures, as well as in situ encapsulation of cells into hydrogels for 3D culture. This review will assess current progress in the preparation of amino acids and peptide-based hydrogels under various kinds of external stimuli, and in situ encapsulation of cells into the hydrogels, with a focus on understanding the associations between their structures, properties, and functions during cell culture, and the remaining challenges in this field. The amino acids and peptide-based hydrogelators with rationally designed structures have promising applications in the fields of regenerative medicine, tissue engineering, and pre-clinical evaluation.
Collapse
Affiliation(s)
- Xiao-Qiu Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road., 200240, Shanghai, China
| | - Chuan-Liang Feng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road., 200240, Shanghai, China
| |
Collapse
|
189
|
Semi-degradable porous poly (vinyl alcohol) hydrogel scaffold for cartilage repair: Evaluation of the initial and cell-cultured tribological properties. J Mech Behav Biomed Mater 2017; 68:163-172. [DOI: 10.1016/j.jmbbm.2017.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/28/2017] [Accepted: 02/02/2017] [Indexed: 11/24/2022]
|
190
|
Nims RJ, Cigan AD, Durney KM, Jones BK, O'Neill JD, Law WSA, Vunjak-Novakovic G, Hung CT, Ateshian GA. * Constrained Cage Culture Improves Engineered Cartilage Functional Properties by Enhancing Collagen Network Stability. Tissue Eng Part A 2017; 23:847-858. [PMID: 28193145 DOI: 10.1089/ten.tea.2016.0467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
When cultured with sufficient nutrient supply, engineered cartilage synthesizes proteoglycans rapidly, producing an osmotic swelling pressure that destabilizes immature collagen and prevents the development of a robust collagen framework, a hallmark of native cartilage. We hypothesized that mechanically constraining the proteoglycan-induced tissue swelling would enhance construct functional properties through the development of a more stable collagen framework. To test this hypothesis, we developed a novel "cage" growth system to mechanically prevent tissue constructs from swelling while ensuring adequate nutrient supply to the growing construct. The effectiveness of constrained culture was examined by testing constructs embedded within two different scaffolds: agarose and cartilage-derived matrix hydrogel (CDMH). Constructs were seeded with immature bovine chondrocytes and cultured under free swelling (FS) conditions for 14 days with transforming growth factor-β before being placed into a constraining cage for the remainder of culture. Controls were cultured under FS conditions throughout. Agarose constructs cultured in cages did not expand after the day 14 caging while FS constructs expanded to 8 × their day 0 weight after 112 days of culture. In addition to the physical differences in growth, by day 56, caged constructs had higher equilibrium (agarose: 639 ± 179 kPa and CDMH: 608 ± 257 kPa) and dynamic compressive moduli (agarose: 3.4 ± 1.0 MPa and CDMH 2.8 ± 1.0 MPa) than FS constructs (agarose: 193 ± 74 kPa and 1.1 ± 0.5 MPa and CDMH: 317 ± 93 kPa and 1.8 ± 1.0 MPa for equilibrium and dynamic properties, respectively). Interestingly, when normalized to final day wet weight, cage and FS constructs did not exhibit differences in proteoglycan or collagen content. However, caged culture enhanced collagen maturation through the increased formation of pyridinoline crosslinks and improved collagen matrix stability as measured by α-chymotrypsin solubility. These findings demonstrate that physically constrained culture of engineered cartilage constructs improves functional properties through improved collagen network maturity and stability. We anticipate that constrained culture may benefit other reported engineered cartilage systems that exhibit a mismatch in proteoglycan and collagen synthesis.
Collapse
Affiliation(s)
- Robert J Nims
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Alexander D Cigan
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Krista M Durney
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Brian K Jones
- 2 Department of Mechanical Engineering, Columbia University , New York, New York
| | - John D O'Neill
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Wing-Sum A Law
- 2 Department of Mechanical Engineering, Columbia University , New York, New York
| | - Gordana Vunjak-Novakovic
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,3 Department of Medicine, Columbia University , New York, New York
| | - Clark T Hung
- 1 Department of Biomedical Engineering, Columbia University , New York, New York
| | - Gerard A Ateshian
- 1 Department of Biomedical Engineering, Columbia University , New York, New York.,2 Department of Mechanical Engineering, Columbia University , New York, New York
| |
Collapse
|
191
|
Kocen R, Gasik M, Gantar A, Novak S. Viscoelastic behaviour of hydrogel-based composites for tissue engineering under mechanical load. ACTA ACUST UNITED AC 2017; 12:025004. [PMID: 28106535 DOI: 10.1088/1748-605x/aa5b00] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Along with biocompatibility, bioinductivity and appropriate biodegradation, mechanical properties are also of crucial importance for tissue engineering scaffolds. Hydrogels, such as gellan gum (GG), are usually soft materials, which may benefit from the incorporation of inorganic particles, e.g. bioactive glass, not only due to the acquired bioactivity, but also due to improved mechanical properties. They exhibit complex viscoelastic properties, which can be evaluated in various ways. In this work, to reliably evaluate the effect of the bioactive glass (BAG) addition on viscoelastic properties of the composite hydrogel, we employed and compared the three most commonly used techniques, analyzing their advantages and limitations: monotonic uniaxial unconfined compression, small amplitude oscillatory shear (SAOS) rheology and dynamic mechanical analysis (DMA). Creep and small amplitude dynamic strain-controlled tests in DMA are suggested as the best ways for the characterization of mechanical properties of hydrogel composites, whereas the SAOS rheology is more useful for studying the hydrogel's processing kinetics, as it does not induce volumetric changes even at very high strains. Overall, the results confirmed a beneficial effect of BAG (nano)particles on the elastic modulus of the GG-BAG composite hydrogel. The Young's modulus of 6.6 ± 0.8 kPa for the GG hydrogel increased by two orders of magnitude after the addition of 2 wt.% BAG particles (500-800 kPa).
Collapse
Affiliation(s)
- Rok Kocen
- Department for Nanostructured Materials, Jožef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia. Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | | | | | | |
Collapse
|
192
|
Rouse CK, Martin AD, Easton CJ, Thordarson P. A Peptide Amphiphile Organogelator of Polar Organic Solvents. Sci Rep 2017; 7:43668. [PMID: 28255169 PMCID: PMC5334642 DOI: 10.1038/srep43668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/27/2017] [Indexed: 11/24/2022] Open
Abstract
A peptide amphiphile is reported, that gelates a range of polar organic solvents including acetonitrile/water, N,N-dimethylformamide and acetone, in a process dictated by β-sheet interactions and facilitated by the presence of an alkyl chain. Similarities with previously reported peptide amphiphile hydrogelators indicate analogous underlying mechanisms of gelation and structure-property relationships, suggesting that peptide amphiphile organogel design may be predictably based on hydrogel precedents.
Collapse
Affiliation(s)
- Charlotte K. Rouse
- Research School of Chemistry, The Australian National University, Canberra, ACT 2601, Australia
| | - Adam D. Martin
- School of Chemistry, The Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Christopher J. Easton
- Research School of Chemistry, The Australian National University, Canberra, ACT 2601, Australia
| | - Pall Thordarson
- School of Chemistry, The Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
193
|
Chan KH, Lee WH, Zhuo S, Ni M. Harnessing supramolecular peptide nanotechnology in biomedical applications. Int J Nanomedicine 2017; 12:1171-1182. [PMID: 28223805 PMCID: PMC5310635 DOI: 10.2147/ijn.s126154] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The harnessing of peptides in biomedical applications is a recent hot topic. This arises mainly from the general biocompatibility of peptides, as well as from the ease of tunability of peptide structure to engineer desired properties. The ease of progression from laboratory testing to clinical trials is evident from the plethora of examples available. In this review, we compare and contrast how three distinct self-assembled peptide nanostructures possess different functions. We have 1) nanofibrils in biomaterials that can interact with cells, 2) nanoparticles that can traverse the bloodstream to deliver its payload and also be bioimaged, and 3) nanotubes that can serve as cross-membrane conduits and as a template for nanowire formation. Through this review, we aim to illustrate how various peptides, in their various self-assembled nanostructures, possess great promise in a wide range of biomedical applications and what more can be expected.
Collapse
Affiliation(s)
| | - Wei Hao Lee
- Department of Chemistry, Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Shuangmu Zhuo
- Fujian Provincial Key Laboratory for Photonics Technology, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, People’s Republic of China
| | - Ming Ni
- Fujian Provincial Key Laboratory for Photonics Technology, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, People’s Republic of China
| |
Collapse
|
194
|
Zhang Y, Song H, Zhang H, Huang P, Liu J, Chu L, Liu J, Wang W, Cheng Z, Kong D. Fine tuning the assembly and gel behaviors of PEGylated polypeptide conjugates by the copolymerization ofl-alanine and γ-benzyl-l-glutamateN-carboxyanhydrides. ACTA ACUST UNITED AC 2017. [DOI: 10.1002/pola.28516] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yumin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Huijuan Song
- Tianjin Key Laboratory of Biomaterial Research; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Hao Zhang
- Ningbo Academy of Agricultural Sciences; Zhejiang 315040 China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Jinjian Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Liping Chu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program; Canary Center at Stanford for Cancer Early Detection, Stanford University; Stanford California 94305 United States
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial Research; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin 300192 China
| |
Collapse
|
195
|
Gao J, Tang C, Elsawy MA, Smith AM, Miller AF, Saiani A. Controlling Self-Assembling Peptide Hydrogel Properties through Network Topology. Biomacromolecules 2017; 18:826-834. [DOI: 10.1021/acs.biomac.6b01693] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jie Gao
- School of Materials, ‡Manchester Institute of Biotechnology, and ∥School of Chemical Engineering and
Analytical Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - Claire Tang
- School of Materials, ‡Manchester Institute of Biotechnology, and ∥School of Chemical Engineering and
Analytical Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - Mohamed A. Elsawy
- School of Materials, ‡Manchester Institute of Biotechnology, and ∥School of Chemical Engineering and
Analytical Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - Andrew M. Smith
- School of Materials, ‡Manchester Institute of Biotechnology, and ∥School of Chemical Engineering and
Analytical Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - Aline F. Miller
- School of Materials, ‡Manchester Institute of Biotechnology, and ∥School of Chemical Engineering and
Analytical Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| | - Alberto Saiani
- School of Materials, ‡Manchester Institute of Biotechnology, and ∥School of Chemical Engineering and
Analytical Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, United Kingdom
| |
Collapse
|
196
|
Xing JZ, Lu L, Unsworth LD, Major PW, Doschak MR, Kaipatur NR. RANKL release from self-assembling nanofiber hydrogels for inducing osteoclastogenesis in vitro. Acta Biomater 2017; 49:306-315. [PMID: 27940164 DOI: 10.1016/j.actbio.2016.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/17/2016] [Accepted: 12/05/2016] [Indexed: 02/01/2023]
Abstract
PURPOSE To develop a nanofiber hydrogel (NF-hydrogel) for sustained and controlled release of the recombinant receptor activator of NF-kB ligand; (RANKL) and to characterize the release kinetics and bioactivity of the released RANKL. METHODS Various concentrations of fluorescently-labelled RANKL protein were added to NF-hydrogels, composed of Acetyl-(Arg-Ala-Asp-Ala)4-CONH2 [(RADA)4] of different concentrations, to investigate the resulting in vitro release rates. The nano-structures of NF-hydrogel, with and without RANKL, were determined using atomic force microscopy (AFM). Released RANKL was further analyzed for changes in secondary and tertiary structure using CD spectroscopy and fluorescent emission spectroscopy, respectively. Bioactivity of released RANKL protein was determined using NFATc1 gene expression and tartrate resistant acid phosphatase (TRAP) activity of osteoclast cells as biomarkers. RESULTS NF-hydrogel concentration dependent sustained release of RANKL protein was measured at concentrations between 0.5 and 2%(w/v). NF-hydrogel at 2%(w/v) concentration exhibited a sustained and slow-release of RANKL protein up to 48h. Secondary and tertiary structure analyses confirmed no changes to the RANKL protein released from NF-hydrogel in comparison to native RANKL. The results of NFATc1 gene mRNA expression and TRAP activities of osteoclast, showed that the release process did not affect the bioactivity of released RANKL. CONCLUSIONS This novel study is the first of its kind to attempt in vitro characterization of NF-hydrogel based delivery of RANKL protein to induce osteoclastogenesis. We have shown the self-assembling NF-hydrogel peptide system is amenable to the sustained and controlled release of RANKL locally; that could in turn increase local concentration of RANKL to induce osteoclastogenesis, for application to the controlled mobilization of tooth movement in orthodontic procedures. STATEMENT OF SIGNIFICANCE Orthodontic tooth movement (OTM) occurs through controlled application of light forces to teeth, facilitating the required changes in the surrounding alveolar bone through the process of bone remodelling. The RANKL system regulates alveolar bone remodelling and controls root resorption during OTM. The use of exogenous RANKL to accelerate OTM has not been attempted to date because large quantities of RANKL for systemic therapy may subsequently cause serious systemic loss of skeletal bone. The controlled and sustained local release of RANKL from a carrier matrix could maximize its therapeutic benefit whilst minimizing systemic side effects. In this study a NF-hydrogel was used for sustained and controlled release of RANKL and the release kinetics and biofunctionality of the released RANKL was characterized. Our results provide fundamental insight for further investigating the role of RANKL NF-hydrogel release systems for inducing osteoclastogenesis in vivo.
Collapse
|
197
|
Wang H, Feng Z, Xu B. D-amino acid-containing supramolecular nanofibers for potential cancer therapeutics. Adv Drug Deliv Rev 2017; 110-111:102-111. [PMID: 27102943 PMCID: PMC5071117 DOI: 10.1016/j.addr.2016.04.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/19/2016] [Accepted: 04/06/2016] [Indexed: 12/30/2022]
Abstract
Nanostructures formed by peptides that self-assemble in water through non-covalent interactions have attracted considerable attention because peptides possess several unique advantages, such as modular design and easiness of synthesis, convenient modification with known functional motifs, good biocompatibility, low immunogenicity and toxicity, inherent biodegradability, and fast responses to a wide range of external stimuli. After about two decades of development, peptide-based supramolecular nanostructures have already shown great potentials in the fields of biomedicine. Among a range of biomedical applications, using such nanostructures for cancer therapy has attracted increased interests since cancer remains the major threat for human health. Comparing with L-peptides, nanostructures containing peptides made of D-amino acid (i.e., D-peptides) bear a unique advantage, biostability (i.e., resistance towards most of endogenous enzymes). The exploration of nanostructures containing D-amino acids, especially their biomedical applications, is still in its infancy. Herein we review the recent progress of D-amino acid-containing supramolecular nanofibers as an emerging class of biomaterials that exhibit unique features for the development of cancer therapeutics. In addition, we give a brief perspective about the challenges and promises in this research direction.
Collapse
Affiliation(s)
- Huaimin Wang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Zhaoqianqi Feng
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA.
| |
Collapse
|
198
|
Chang B, Ahuja N, Ma C, Liu X. Injectable scaffolds: Preparation and application in dental and craniofacial regeneration. MATERIALS SCIENCE & ENGINEERING. R, REPORTS : A REVIEW JOURNAL 2017; 111:1-26. [PMID: 28649171 PMCID: PMC5478172 DOI: 10.1016/j.mser.2016.11.001] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Injectable scaffolds are appealing for tissue regeneration because they offer many advantages over pre-formed scaffolds. This article provides a comprehensive review of the injectable scaffolds currently being investigated for dental and craniofacial tissue regeneration. First, we provide an overview of injectable scaffolding materials, including natural, synthetic, and composite biomaterials. Next, we discuss a variety of characteristic parameters and gelation mechanisms of the injectable scaffolds. The advanced injectable scaffolding systems developed in recent years are then illustrated. Furthermore, we summarize the applications of the injectable scaffolds for the regeneration of dental and craniofacial tissues that include pulp, dentin, periodontal ligament, temporomandibular joint, and alveolar bone. Finally, our perspectives on the injectable scaffolds for dental and craniofacial tissue regeneration are offered as signposts for the future advancement of this field.
Collapse
Affiliation(s)
- Bei Chang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Neelam Ahuja
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Chi Ma
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| |
Collapse
|
199
|
Wang Y, Xiao Y, Gao G, Chen J, Hou R, Wang Q, Liu L, Fu J. Conductive graphene oxide hydrogels reduced and bridged by l-cysteine to support cell adhesion and growth. J Mater Chem B 2017; 5:511-516. [DOI: 10.1039/c6tb02333a] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
l-Cysteine reduces and bridges graphene oxide into a network, yielding conductive hydrogels nicely supporting cell adhesion and growth.
Collapse
Affiliation(s)
- Yifan Wang
- School of Materials Science and Engineering
- Shanghai University
- Shanghai
- China
- Cixi Institute of Biomedical Engineering
| | - Ying Xiao
- Cixi Institute of Biomedical Engineering
- Ningbo Institute of Materials Technology & Engineering
- Chinese Academy of Sciences
- Ningbo 315300
- China
| | - Guorong Gao
- Cixi Institute of Biomedical Engineering
- Ningbo Institute of Materials Technology & Engineering
- Chinese Academy of Sciences
- Ningbo 315300
- China
| | - Jing Chen
- Cixi Institute of Biomedical Engineering
- Ningbo Institute of Materials Technology & Engineering
- Chinese Academy of Sciences
- Ningbo 315300
- China
| | - Ruixia Hou
- Cixi Institute of Biomedical Engineering
- Ningbo Institute of Materials Technology & Engineering
- Chinese Academy of Sciences
- Ningbo 315300
- China
| | - Qiang Wang
- Cixi Institute of Biomedical Engineering
- Ningbo Institute of Materials Technology & Engineering
- Chinese Academy of Sciences
- Ningbo 315300
- China
| | - Li Liu
- School of Materials Science and Engineering
- Shanghai University
- Shanghai
- China
| | - Jun Fu
- Cixi Institute of Biomedical Engineering
- Ningbo Institute of Materials Technology & Engineering
- Chinese Academy of Sciences
- Ningbo 315300
- China
| |
Collapse
|
200
|
Chuah YJ, Peck Y, Lau JEJ, Hee HT, Wang DA. Hydrogel based cartilaginous tissue regeneration: recent insights and technologies. Biomater Sci 2017; 5:613-631. [DOI: 10.1039/c6bm00863a] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Hydrogel based technologies has been extensively employed in both exploratory research and clinical applications to address numerous existing challenges in the regeneration of articular cartilage and intervertebral disc.
Collapse
Affiliation(s)
- Yon Jin Chuah
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637459
- Singapore
| | - Yvonne Peck
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637459
- Singapore
| | - Jia En Josias Lau
- School of Chemical & Life Sciences
- Singapore Polytechnic
- Singapore 139651
- Singapore
| | - Hwan Tak Hee
- Lee Kong Chian School of Medicine
- Nanyang Technological University
- Singapore 636921
- Singapore
- Pinnacle Spine & Scoliosis Centre
| | - Dong-An Wang
- School of Chemical and Biomedical Engineering
- Nanyang Technological University
- Singapore 637459
- Singapore
| |
Collapse
|