1
|
Crump KB, Kanelis E, Segarra-Queralt M, Pascuet-Fontanet A, Bermudez-Lekerika P, Alminnawi A, Geris L, Alexopoulos LG, Noailly J, Gantenbein B. TNF induces catabolism in human cartilaginous endplate cells in 3D agarose culture under dynamic compression. Sci Rep 2025; 15:15849. [PMID: 40328789 PMCID: PMC12056083 DOI: 10.1038/s41598-025-00538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Intervertebral disc (IVD) degeneration is the leading cause of low back pain in young adults, and the cartilaginous endplate (CEP) is likely to play a key role in early IVD degeneration. To elucidate the effects of pro-inflammatory cytokines on the mechanobiology of the CEP, human CEP cells were seeded into 2% agarose, dynamically compressed up to 7%, and stimulated with tumor necrosis factor (TNF). It was hypothesized that dynamic compression would be sufficient to induce anabolism, while stimulation with TNF would induce catabolism. TNF was sufficient to induce a catabolic, time-dependent response in human CEP cells through downregulation of anabolic gene expression and increased secretion of pro-inflammatory proteins associated with herniated discs, bacteria inhibition, and pain. However, 7% strain or scaffold material, agarose, may not lead to full activation of integrins and downregulation of pro-inflammatory pathways, demonstrated in part through the unchanged gene expression of integrin subunits α5 and β1.
Collapse
Affiliation(s)
- Katherine B Crump
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012, Bern, Switzerland
| | - Exarchos Kanelis
- School of Mechanical Engineering, National Technical University of Athens, 15772, Zografou, Greece
- Protavio Ltd, 15341, Agia Paraskevi, Greece
| | | | | | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012, Bern, Switzerland
| | - Ahmad Alminnawi
- GIGA In Silico Medicine, University of Liège, Liège, 4000, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Liesbet Geris
- GIGA In Silico Medicine, University of Liège, Liège, 4000, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Leonidas G Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, 15772, Zografou, Greece
- Protavio Ltd, 15341, Agia Paraskevi, Greece
| | - Jérôme Noailly
- BCN Medtech, Universitat Pompeu Fabra, 08018, Barcelona, Spain
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland.
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Faculty of Medicine, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
2
|
Mukundan LM, Rajasekaran R, Das S, Seesala VS, Ganguly D, Kumar N, Dhara S, Chattopadhyay S. Tailoring of agarose hydrogel to modulate its 3D bioprintability and mechanical properties for stem cell mediated bone tissue engineering. Int J Biol Macromol 2025; 309:142795. [PMID: 40185455 DOI: 10.1016/j.ijbiomac.2025.142795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 03/17/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The intense gelling characteristics and viscosity constraints of agarose limit its utility as a sole ink material in 3D printing. This study presents the development of agarose bioink designed for cell-laden printing, featuring controlled printability, exceptional stiffness, and cell-responsiveness achieved via the insertion of photochemically reactive methacrylate groups. This chemical modification transforms the dense agarose network into a thinner structure, effecting a gentle thermogelling property that enhances the printability and facile cell encapsulation. Herein we examine the interplay between the degree of substitution and concentration variations to determine the optimal hydrogel composition. The best bioink composition possessed a lower shear modulus (storage modulus G' = 11.6 Pa) at 37 °C, assuring better bioprintability, while it possessed a Young's modulus of 1.4 ± 0.10 MPa in the crosslinked state, which is the highest reported in the natural single-matrix hydrogel systems. Studies with mesenchymal stem cells (MSC) confirmed that it is a good cell encapsulation matrix, achieving 111 % cell viability at 72 h. The bioprinted constructs promoted the osteogenic differentiation of MSC, as evidenced by mineralization and secretion of bone-related matrix. The gene expression analysis indicated that osteogenic marker expressions exhibited at least a two-fold increase on day 14 relative to the control group.
Collapse
Affiliation(s)
- Lakshmi M Mukundan
- Rubber Technology Center, Indian Institute of Technology Kharagpur, West Bengal 721302, India; School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Ragavi Rajasekaran
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Samir Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - V S Seesala
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Debabrata Ganguly
- Rubber Technology Center, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Nikhil Kumar
- Rubber Technology Center, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Santanu Dhara
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Santanu Chattopadhyay
- Rubber Technology Center, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
3
|
Feng Y, Jiang Z, Chen C, Hu L, Jiang Q, Wang Y, Cheng Z, Wang F, Yang G, Wang Y. Laminin expression profiles of osteogenic-and chondrogenic-induced dECM sheets. BIOMATERIALS ADVANCES 2025; 169:214127. [PMID: 39637724 DOI: 10.1016/j.bioadv.2024.214127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Decellularized extracellular matrix sheets (dECMSs) produced by stem cells have attracted attention because they preserve the natural biological activity of the ECM to direct lineage-specific differentiation with less immunogenicity. As a core ECM protein, laminin modulates cellular phenotype and differentiation. Nevertheless, no studies thus far have explored the distribution and abundance of laminins in diverse dECMSs. Herein, we first compared the differential expression of laminins among dECMSs in osteogenic-induced medium (OI-dECMS), chondrogenic-induced medium (CI-dECMS), and standard medium (dECMS), employing a defined mass spectrometry (MS)-based proteomic analysis. In vitro, dECMSs were verified to be successfully decellularized. Cluster analysis identified a marked fluctuation in the expression of 7 laminins and 17 laminin-associated proteins in OI-dECMS vs dECMS and CI-dECMS vs dECMS. Two significantly changed pathways were selected from the KEGG pathway enrichment analysis: the FAK/ERK pathway and the PI3K/AKT pathway. Moreover, Alkaline Phosphatase (ALP) activity, Alcian blue staining, and RT-qPCR results for recellularization showed that CI-dECMS promotes chondrogenesis while OI-dECMS inhibits osteogenesis compared with dECMS. In vivo experiments were conducted to implant dECMSs in a rat osteochondral defect, demonstrating that dECMS and CI-dECMS promoted bone and cartilage repair. Furthermore, the inhibitory analysis was performed to verify the function of specific laminin isoforms modulating osteogenesis and chondrogenesis, which might be related to FAK/ERK and PI3K/AKT pathways. In summary, this study constructed dECMS, OI-dECMS, and CI-dECMS and uncovered the internal comprehensive molecular regulatory network centralized by laminins, thus proposing a biomimetic substitute for bone and cartilage regeneration.
Collapse
Affiliation(s)
- Yuting Feng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Chaozhen Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Ling Hu
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou 311100, China
| | - Qifeng Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Yuchen Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhenxuan Cheng
- Affiliated Stomatology Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310059, China
| | - Fang Wang
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou 311100, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
4
|
Huang Y, Peng S, Chen Y, Chu B. Agarose Hydrogels for Bone Tissue Engineering, from Injectables to Bioprinting. Gels 2025; 11:255. [PMID: 40277691 PMCID: PMC12027395 DOI: 10.3390/gels11040255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
A great interest in agarose, with many health-promoting and gel properties, has been registered, especially in the field of bone regeneration and repair. Agarose and its major bioactive compounds are involved in biological activities such as inflammation, cell adhesion and proliferation, and the promotion of tissue repair. Due to its unique physical properties like gelation and solubility, agarose is increasingly utilized in the medical industry. The aim of this review is to present an overview of the applications of agarose hydrogels in bone tissue engineering, introducing agarose and its modified products as innovative solutions for bone regeneration. Additionally, the injectability of agarose hydrogels and their applications in bioprinting are also summarized. Data indicate that agarose will play an increasing role in current and future global medical sectors.
Collapse
Affiliation(s)
- Yibin Huang
- School of Materials Science and Engineering, Xiamen University of Technology, Xiamen 361024, China
| | - Siyuan Peng
- School of Materials Science and Engineering, Xiamen University of Technology, Xiamen 361024, China
| | - Yifan Chen
- School of Materials Science and Engineering, Xiamen University of Technology, Xiamen 361024, China
| | - Bin Chu
- School of Materials Science and Engineering, Xiamen University of Technology, Xiamen 361024, China
- Key Laboratory of Biomedical Materials and Implant Devices, Research Institute, Tsinghua University, Shenzhen 518057, China
| |
Collapse
|
5
|
Piazza F, Ravaglia B, Caporale A, Svetić A, Parisse P, Asaro F, Grassi G, Secco L, Sgarra R, Marsich E, Donati I, Sacco P. Elucidating the unexpected cell adhesive properties of agarose substrates. The effect of mechanics, fetal bovine serum and specific peptide sequences. Acta Biomater 2024; 189:286-297. [PMID: 39357634 DOI: 10.1016/j.actbio.2024.09.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
2D agarose substrates have recently been surprisingly shown to be permissive for cell adhesion, depending on their mechanics and the use of the adhesive proteins of fetal bovine serum (FBS) in the cell culture medium. Here, we elucidate how the cells exhibit two anchoring mechanisms depending on the amount of FBS. Under low FBS conditions, the cells recognize the surface-coupled adhesive sequences of fibronectin via the binding of the heterodimer α5β1 integrin. Functionality of the actomyosin axis and mechanoactivation of focal adhesion kinase (FAK) are essential for the stretching of the protein, thereby accessing the "synergy" PPSRN site and enhancing cell adhesion in combination with the downstream RGD motif. Under high FBS conditions, the specific peptide sequences are much less relevant as the adsorbed serum proteins conceal the coupled fibronectin and the cells recognize the adhesive protein vitronectin, which is constitutively present in FBS, via the binding of the heterodimer αvβ3 integrin. Similarly, the intracellular tension and FAK activity are decisive, which collectively indicate that the cells stretch the partially cryptic RGD site of vitronectin and thus make it more accessible for integrin binding. Both anchoring mechanisms only work properly if the agarose substrate is mechanically compliant in terms of linear stress-strain response, unraveling a critical balance between the mechanics of the agarose substrate and the presentation of the adhesive peptides. STATEMENT OF SIGNIFICANCE: In the context of biomaterial design, agarose hydrogels are known to lack intrinsic cell-adhesive peptide motifs and are therefore commonly used for the development of non-permissive 2D substrates. However, we unexpectedly found that agarose hydrogels can become permissive substrates for cell adhesion, depending on a compliant mechanical response of the substrate and the use of fetal bovine serum (FBS) as protein reservoir in the cell culture medium. We describe here two anchoring mechanisms that cells harness to adhere to agarose substrates, depending on the amount of FBS. Our results will have a major impact on the field of mechanobiology and shed light on the central role of FBS as a natural source of adhesive proteins that could promote cell anchoring.
Collapse
Affiliation(s)
- Francesco Piazza
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Beatrice Ravaglia
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Andrea Caporale
- CNR, Institute of Crystallography, I-34149 Trieste, Italy; CIRPeB, Research Centre on Bioactive Peptides "Carlo Pedone", University of Naples "Federico II", I-80134 Napoli, Italy
| | - Ana Svetić
- Department of Physics, University of Trieste, Via Alfonso Valerio 2, I-34127 Trieste, Italy; Elettra Sincrotrone Trieste S.C.p.A., s.s. 14km 1635 in Area Science Park, I-34149 Trieste, Italy
| | - Pietro Parisse
- Elettra Sincrotrone Trieste S.C.p.A., s.s. 14km 1635 in Area Science Park, I-34149 Trieste, Italy; Istituto Officina dei Materiali (CNR-IOM), Consiglio Nazionale delle Ricerche, s.s. 14km 1635 in Area Science Park, I-34149 Trieste, Italy
| | - Fioretta Asaro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Licio Giorgieri 1, I-34127 Trieste, Italy
| | - Gabriele Grassi
- Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 447, I-34129 Trieste, Italy
| | - Luca Secco
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Eleonora Marsich
- Department of Medical, Surgical and Health Science, University of Trieste, Strada di Fiume 447, I-34129 Trieste, Italy
| | - Ivan Donati
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy
| | - Pasquale Sacco
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, I-34127 Trieste, Italy.
| |
Collapse
|
6
|
Jafari A. Advancements in self-assembling peptides: Bridging gaps in 3D cell culture and electronic device fabrication. J Biomater Appl 2024; 38:1013-1035. [PMID: 38502905 PMCID: PMC11055414 DOI: 10.1177/08853282241240139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Self-assembling peptides (SAPs) show promise in creating synthetic microenvironments that regulate cellular function and tissue repair. Also, the precise π-π interactions and hydrogen bonding within self-assembled peptide structures enable the creation of quantum confined structures, leading to reduced band gaps and the emergence of semiconductor properties within the superstructures. This review emphasizes the need for standardized 3D cell culture methods and electronic devices based on SAPs for monitoring cell communication and controlling cell surface morphology. Additionally, the gap in understanding the relationship between SAP peptide sequences and nanostructures is highlighted, underscoring the importance of optimizing peptide deposition parameters, which affect charge transport and bioactivity due to varying morphologies. The potential of peptide nanofibers as extracellular matrix mimics and the introduction of the zone casting method for improved film deposition are discussed within this review, aiming to bridge knowledge gaps and offer insights into fields like tissue engineering and materials science, with the potential for groundbreaking applications at the interface of biology and materials engineering.
Collapse
Affiliation(s)
- Azadeh Jafari
- Faculty of Applied Sciences, Simon Fraser University, Surrey, BC, Canada
| |
Collapse
|
7
|
Yamada Y. Characterization of Novel Cell-Adhesive Peptides for Biomaterial Development. Biol Pharm Bull 2024; 47:1072-1078. [PMID: 38825460 DOI: 10.1248/bpb.b24-00030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
In previous studies, my group developed cell-adhesive peptide-polysaccharide complexes as biomaterials for tissue engineering. Having a wide variety of cell-adhesive peptides is important as the biological functions of peptide-polysaccharide complexes are highly dependent on the biological activity of peptides. This paper reviews the biological activities of two types of recently characterized cell-adhesive peptides. The first is peptides rich in basic amino acids originating from octaarginine. We analyzed the relationships between the amino acid composition of basic peptides and cell adhesion, elongation, and proliferation and identified the most suitable peptide for cell culture. The second was arginine-glycine-aspartic acid (RGD)-containing peptides that promote the adhesion of induced pluripotent stem cells (iPSCs). We identified the RGD-surrounding sequences necessary for iPSC adhesion, clarified the underlying mechanism, and improved cell adhesion by modifying the structure-activity relationships. The novel cell-adhesive peptides identified in our previous studies may aid in the development of novel peptide-based biomaterials.
Collapse
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
8
|
Li C, Holman JB, Shi Z, Qiu B, Ding W. On-chip modeling of tumor evolution: Advances, challenges and opportunities. Mater Today Bio 2023; 21:100724. [PMID: 37483380 PMCID: PMC10359640 DOI: 10.1016/j.mtbio.2023.100724] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Tumor evolution is the accumulation of various tumor cell behaviors from tumorigenesis to tumor metastasis and is regulated by the tumor microenvironment (TME). However, the mechanism of solid tumor progression has not been completely elucidated, and thus, the development of tumor therapy is still limited. Recently, Tumor chips constructed by culturing tumor cells and stromal cells on microfluidic chips have demonstrated great potential in modeling solid tumors and visualizing tumor cell behaviors to exploit tumor progression. Herein, we review the methods of developing engineered solid tumors on microfluidic chips in terms of tumor types, cell resources and patterns, the extracellular matrix and the components of the TME, and summarize the recent advances of microfluidic chips in demonstrating tumor cell behaviors, including proliferation, epithelial-to-mesenchymal transition, migration, intravasation, extravasation and immune escape of tumor cells. We also outline the combination of tumor organoids and microfluidic chips to elaborate tumor organoid-on-a-chip platforms, as well as the practical limitations that must be overcome.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Joseph Benjamin Holman
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Weiping Ding
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
9
|
Yu X, Wang L, He W. Cytophilic Agarose-Epoxide-Amine Cryogels Engineered with Granulated Microstructures. ACS APPLIED BIO MATERIALS 2023; 6:694-702. [PMID: 36695539 DOI: 10.1021/acsabm.2c00938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Inherent cytophobicity of agarose limits its direct use for the growth of anchorage-dependent cells. Here, we report a simple strategy allowing the development of agarose-based hydrogels entailed with both cytophilicity and microstructured morphology. Through the reaction of water-soluble 1,4-butanediol diglycidyl ether (BDDE) with trifunctional polyetheramine Jeffamine T403 in agarose solution followed by cryogelation of the mixtures, a series of macroporous agarose-epoxide-amine cryogels were prepared readily. Results from fluorescent labeling and energy-dispersive X-ray elemental mapping showed the formation of granulated microstructures in the cryogels. Such features closely correlated to the phase separation of BDDE-T403 polymers within the agarose matrix. Cytophilicity of the microstructured cryogels due to the integrated amine moieties was demonstrated through the adhesion of fibroblasts. Functional enrichment of the cryogels was further highlighted by leveraging the granulates as micro-reservoirs for polyphenol proanthocyanidin to enable antioxidation and protection of fibroblasts from H2O2-induced cytotoxic effect in vitro.
Collapse
Affiliation(s)
- Xueying Yu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, Liaoning116024, China.,School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning116024, China
| | - Liwei Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, Liaoning116024, China.,School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning116024, China
| | - Wei He
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, Liaoning116024, China.,School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning116024, China
| |
Collapse
|
10
|
Hajiabbas M, D'Agostino C, Simińska-Stanny J, Tran SD, Shavandi A, Delporte C. Bioengineering in salivary gland regeneration. J Biomed Sci 2022; 29:35. [PMID: 35668440 PMCID: PMC9172163 DOI: 10.1186/s12929-022-00819-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Salivary gland (SG) dysfunction impairs the life quality of many patients, such as patients with radiation therapy for head and neck cancer and patients with Sjögren’s syndrome. Multiple SG engineering strategies have been considered for SG regeneration, repair, or whole organ replacement. An in-depth understanding of the development and differentiation of epithelial stem and progenitor cells niche during SG branching morphogenesis and signaling pathways involved in cell–cell communication constitute a prerequisite to the development of suitable bioengineering solutions. This review summarizes the essential bioengineering features to be considered to fabricate an engineered functional SG model using various cell types, biomaterials, active agents, and matrix fabrication methods. Furthermore, recent innovative and promising approaches to engineering SG models are described. Finally, this review discusses the different challenges and future perspectives in SG bioengineering.
Collapse
Affiliation(s)
- Maryam Hajiabbas
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium
| | - Claudia D'Agostino
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium
| | - Julia Simińska-Stanny
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland.,3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium.
| |
Collapse
|
11
|
Yamada Y, Onda T, Hamada K, Kikkawa Y, Nomizu M. Octa-arginine and Octa-lysine Promote Cell Adhesion through Heparan Sulfate Proteoglycans and Integrins. Biol Pharm Bull 2022; 45:207-212. [PMID: 35110508 DOI: 10.1248/bpb.b21-00791] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Octa-arginine (R8) has been extensively studied as a cell-penetrating peptide. R8 binds to diverse transmembrane heparan sulfate proteoglycans (HSPGs), including syndecans, and is internalized by cells. R8 is also reported to bind to integrin β1. In this study, we evaluated the biological activities of R8 and octa-lysine (K8), a peptide similar to R8, with a focus on cell adhesion. R8 and K8 were immobilized on aldehyde-agarose matrices via covalent conjugation, and the effect of these peptides on cell attachment, spreading, and proliferation was examined using human dermal fibroblasts. The results indicated that R8- and K8-matrices mediate cell adhesion mainly via HSPGs. Moreover, R8- and K8-matrices interacted with integrin β1 and promote cell spreading and proliferation. These results are useful for further understanding of the R8-membrane interactions and the cellular uptake mechanisms. In addition, the R8- and K8-matrices may potentially be used as a multi-functional biomaterial to promote cell adhesion, spreading, and proliferation.
Collapse
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Toru Onda
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
12
|
Alheib O, da Silva LP, Caballero D, Pires RA, Kundu SC, Correlo VM, Reis RL. Micropatterned gellan gum-based hydrogels tailored with laminin-derived peptides for skeletal muscle tissue engineering. Biomaterials 2021; 279:121217. [PMID: 34781243 DOI: 10.1016/j.biomaterials.2021.121217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 01/13/2023]
Abstract
The efficacy of current therapies for skeletal muscle disorders/injuries are limited urging the need for new treatments. Skeletal muscle tissue engineered platforms represent a promising tool to shed light on the pathophysiology of skeletal muscle disorders/injuries and to investigate the efficacy of new therapies. Herein, we developed a skeletal muscle platform composed of aligned and differentiated myoblasts on micropatterned gellan gum (GG)-based hydrogels tailored with a laminin-derived peptide. To this aim, the binding of murine skeletal muscle cells (C2C12) to different laminin-derived peptides (CIKVAVS (V), KNRLTIELEVRTC (T), and RKRLQVQLSIRTC (Q)) and the binding of laminin-derived peptides to chemically functionalized GG was studied. C2C12-binding to peptide V, T and Q was 10%, 48% and 25%, whereas the peptide tethering to GG was 60%, 40% and 31%, respectively. Peptide-biofunctionalized hydrogels prepared with different polymer content showed different mechanics and peptide exposure at hydrogel surface. Cellular adhesion was detected in all hydrogel formulations, but spreading and differentiation was only promoted in peptide Q-biofunctionalized hydrogels and preferably in stiffer hydrogels. Myoblast alignment was promoted in micropatterned hydrogel surfaces. Overall, the engineered skeletal muscle herein proposed can be further explored as a platform to better understand skeletal muscle disorders/injuries and to screen new therapies.
Collapse
Affiliation(s)
- Omar Alheib
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Lucilia P da Silva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| | - David Caballero
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Ricardo A Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Vitor M Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| |
Collapse
|
13
|
Kapr J, Petersilie L, Distler T, Lauria I, Bendt F, Sauter CM, Boccaccini AR, Rose CR, Fritsche E. Human Induced Pluripotent Stem Cell-Derived Neural Progenitor Cells Produce Distinct Neural 3D In Vitro Models Depending on Alginate/Gellan Gum/Laminin Hydrogel Blend Properties. Adv Healthc Mater 2021; 10:e2100131. [PMID: 34197049 PMCID: PMC11468953 DOI: 10.1002/adhm.202100131] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/01/2021] [Indexed: 12/12/2022]
Abstract
Stable and predictive neural cell culture models are a necessary premise for many research fields. However, conventional 2D models lack 3D cell-material/-cell interactions and hence do not reflect the complexity of the in vivo situation properly. Here two alginate/gellan gum/laminin (ALG/GG/LAM) hydrogel blends are presented for the fabrication of human induced pluripotent stem cell (hiPSC)-based 3D neural models. For hydrogel embedding, hiPSC-derived neural progenitor cells (hiNPCs) are used either directly or after 3D neural pre-differentiation. It is shown that stiffness and stress relaxation of the gel blends, as well as the cell differentiation strategy influence 3D model development. The embedded hiNPCs differentiate into neurons and astrocytes within the gel blends and display spontaneous intracellular calcium signals. Two fit-for-purpose models valuable for i) applications requiring a high degree of complexity, but less throughput, such as disease modeling and long-term exposure studies and ii) higher throughput applications, such as acute exposures or substance screenings are proposed. Due to their wide range of applications, adjustability, and printing capabilities, the ALG/GG/LAM based 3D neural models are of great potential for 3D neural modeling in the future.
Collapse
Affiliation(s)
- Julia Kapr
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Laura Petersilie
- Institute of NeurobiologyHeinrich Heine UniversityDüsseldorf40225Germany
| | - Thomas Distler
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐University Erlangen‐NurembergErlangen91054Germany
| | - Ines Lauria
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Farina Bendt
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Clemens M. Sauter
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
| | - Aldo R. Boccaccini
- Institute of BiomaterialsDepartment of Materials Science and EngineeringFriedrich‐Alexander‐University Erlangen‐NurembergErlangen91054Germany
| | - Christine R. Rose
- Institute of NeurobiologyHeinrich Heine UniversityDüsseldorf40225Germany
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental MedicineDüsseldorf40225Germany
- Medical FacultyHeinrich Heine UniversityDüsseldorf40225Germany
| |
Collapse
|
14
|
Abstract
Defined by its potential for self-renewal, differentiation and tumorigenicity, cancer stem cells (CSCs) are considered responsible for drug resistance and relapse. To understand the behavior of CSC, the effects of the microenvironment in each tissue are a matter of great concerns for scientists in cancer biology. However, there are many complicated obstacles in the mimicking the microenvironment of CSCs even with current advanced technology. In this context, novel biomaterials have widely been assessed as in vitro platforms for their ability to mimic cancer microenvironment. These efforts should be successful to identify and characterize various CSCs specific in each type of cancer. Therefore, extracellular matrix scaffolds made of biomaterial will modulate the interactions and facilitate the investigation of CSC associated with biological phenomena simplifying the complexity of the microenvironment. In this review, we summarize latest advances in biomaterial scaffolds, which are exploited to mimic CSC microenvironment, and their chemical and biological requirements with discussion. The discussion includes the possible effects on both cells in tumors and microenvironment to propose what the critical factors are in controlling the CSC microenvironment focusing the future investigation. Our insights on their availability in drug screening will also follow the discussion.
Collapse
|
15
|
Yamada Y, Yoshida C, Hamada K, Kikkawa Y, Nomizu M. Development of Three-Dimensional Cell Culture Scaffolds Using Laminin Peptide-Conjugated Agarose Microgels. Biomacromolecules 2020; 21:3765-3771. [DOI: 10.1021/acs.biomac.0c00871] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chihiro Yoshida
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
16
|
Mao Z, Bi X, Ye F, Shu X, Sun L, Guan J, Ritchie RO, Wu S. Controlled Cryogelation and Catalytic Cross-Linking Yields Highly Elastic and Robust Silk Fibroin Scaffolds. ACS Biomater Sci Eng 2020; 6:4512-4522. [PMID: 33455190 DOI: 10.1021/acsbiomaterials.0c00752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Silk biomaterials with tunable mechanical properties and biological properties are of special importance for tissue engineering. Here, we fabricated silk fibroin (SF, from Bombyx mori silk) scaffolds from cryogelation under controlled temperature and catalytic cross-linking conditions. Structurally, the cryogelled scaffolds demonstrated a greater β-sheet content but significantly smaller β-sheet domains compared to that without chemical cross-linking and catalyst. Mechanically, the cryogelled scaffolds were softer and highly elastic under tension and compression. The 120% tensile elongation and >85% recoverable compressive strain were among the best properties reported for SF scaffolds. Cyclic compression tests proved the robustness of such scaffolds to resist fatigue. The mechanical properties, as well as the degradation rate of the scaffolds, can be fine-tuned by varying the concentrations of the catalyst and the cross-linker. For biological responses, in vitro rat bone mesenchymal stem cell (rBMSC) culture studies demonstrated that cryogelled SF scaffolds supported better cell attachment and proliferation than the routine freeze-thawed scaffolds. The in vivo subcutaneous implantation results showed excellent histocompatibility and tissue ingrowth for the cryogelled SF scaffolds. This straightforward approach of enhanced elasticity of SF scaffolds and fine-tunability in mechanical performances, suggests a promising strategy to develop novel SF biomaterials for soft tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zhinan Mao
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| | - Xuewei Bi
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beijing 100083, China
| | - Fan Ye
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| | - Xiong Shu
- Beijing Research Institute of Traumatology & Orthopaedics, Beijing 100035, China
| | - Lei Sun
- Beijing Research Institute of Traumatology & Orthopaedics, Beijing 100035, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beijing 100083, China
| | - Robert O Ritchie
- Department of Materials Science & Engineering, University of California, Berkeley, California 94720, United States
| | - Sujun Wu
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
17
|
Jose G, Shalumon K, Chen JP. Natural Polymers Based Hydrogels for Cell Culture Applications. Curr Med Chem 2020; 27:2734-2776. [PMID: 31480996 DOI: 10.2174/0929867326666190903113004] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
Abstract
It is well known that the extracellular matrix (ECM) plays a vital role in the growth, survival
and differentiation of cells. Though two-dimensional (2D) materials are generally used as substrates for
the standard in vitro experiments, their mechanical, structural, and compositional characteristics can
alter cell functions drastically. Many scientists reported that cells behave more natively when cultured
in three-dimensional (3D) environments than on 2D substrates, due to the more in vivo-like 3D cell
culture environment that can better mimic the biochemical and mechanical properties of the ECM. In
this regard, water-swollen network polymer-based materials called hydrogels are highly attractive for
developing 3D ECM analogs due to their biocompatibility and hydrophilicity. Since hydrogels can be
tuned and altered systematically, these materials can function actively in a defined culture medium to
support long-term self-renewal of various cells. The physico-chemical and biological properties of the
materials used for developing hydrogel should be tunable in accordance with culture needs. Various
types of hydrogels derived either from natural or synthetic origins are currently being used for cell culture
applications. In this review, we present an overview of various hydrogels based on natural polymers
that can be used for cell culture, irrespective of types of applications. We also explain how each
hydrogel is made, its source, pros and cons in biological applications with a special focus on regenerative
engineering.
Collapse
Affiliation(s)
- Gils Jose
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - K.T. Shalumon
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| |
Collapse
|
18
|
Cambria E, Brunner S, Heusser S, Fisch P, Hitzl W, Ferguson SJ, Wuertz-Kozak K. Cell-Laden Agarose-Collagen Composite Hydrogels for Mechanotransduction Studies. Front Bioeng Biotechnol 2020; 8:346. [PMID: 32373605 PMCID: PMC7186378 DOI: 10.3389/fbioe.2020.00346] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
The increasing investigation of cellular mechanotransduction mechanisms requires biomaterials combining biofunctionality and suitable mechanical properties. Agarose is a standard biomaterial for cartilage and intervertebral disc mechanobiology studies, but lacks adhesion motifs and the necessary cell-matrix interaction for mechanotransduction. Here, collagen type I was blended at two concentrations (2 and 4.5 mg/mL) with agarose 2% wt/vol. The composite hydrogels were characterized in terms of structural homogeneity, rheological properties and size stability. Nucleus pulposus (NP) cell viability, proliferation, morphology, gene expression, GAG production, adhesion and mechanotransduction ability were further tested. Blended hydrogels presented a homogenous network of the two polymers. While the addition of 4.5 mg/mL collagen significantly decreased the storage modulus and increased the loss modulus of the gels, blended gels containing 2 mg/mL collagen displayed similar mechanical properties to agarose. Hydrogel size was conserved over 21 days for all agarose-based gels. Embedded cells were viable (>80%) and presented reduced proliferation and a round morphology typical of NP cells in vivo. Gene expression of collagen types I and II and aggrecan significantly increased in blended hydrogels from day 1 to 7, further resulting in a significantly superior GAG/DNA ratio compared to agarose gels at day 7. Agarose-collagen hydrogels not only promoted cell adhesion, contrary to agarose gels, but also showed a 5.36-fold higher focal adhesion kinase phosphorylation (pFAK/β-tubulin) when not compressed, and increased pFAK/FAK values 10 min after compression. Agarose-collagen thus outperforms agarose, mimics native tissues constituted of non-fibrillar matrix and collagens, and allows exploring complex loading in a highly reproducible system.
Collapse
Affiliation(s)
- Elena Cambria
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Silvio Brunner
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Sally Heusser
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Philipp Fisch
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Wolfgang Hitzl
- Research Office (Biostatistics), Paracelsus Medical University, Salzburg, Austria.,Department of Ophthalmology and Optometry, Paracelsus Medical University, Salzburg, Austria.,Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | | | - Karin Wuertz-Kozak
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.,Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, United States.,Spine Center, Schön Klinik München Harlaching, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Private Medical University Salzburg (Austria), Munich, Germany
| |
Collapse
|
19
|
Barros D, Amaral IF, Pêgo AP. Laminin-Inspired Cell-Instructive Microenvironments for Neural Stem Cells. Biomacromolecules 2019; 21:276-293. [PMID: 31789020 DOI: 10.1021/acs.biomac.9b01319] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Laminin is a heterotrimeric glycoprotein with a key role in the formation and maintenance of the basement membrane architecture and properties, as well as on the modulation of several biological functions, including cell adhesion, migration, differentiation and matrix-mediated signaling. In the central nervous system (CNS), laminin is differentially expressed during development and homeostasis, with an impact on the modulation of cell function and fate. Within neurogenic niches, laminin is one of the most important and well described extracellular matrix (ECM) proteins. Specifically, efforts have been made to understand laminin assembly, domain architecture, and interaction of its different bioactive domains with cell surface receptors, soluble signaling molecules, and ECM proteins, to gain insight into the role of this ECM protein and its receptors on the modulation of neurogenesis, both in homeostasis and during repair. This is also expected to provide a rational basis for the design of biomaterial-based matrices mirroring the biological properties of the basement membrane of neural stem cell niches, for application in neural tissue repair and cell transplantation. This review provides a general overview of laminin structure and domain architecture, as well as the main biological functions mediated by this heterotrimeric glycoprotein. The expression and distribution of laminin in the CNS and, more specifically, its role within adult neural stem cell niches is summarized. Additionally, a detailed overview on the use of full-length laminin and laminin derived peptide/recombinant laminin fragments for the development of hydrogels for mimicking the neurogenic niche microenvironment is given. Finally, the main challenges associated with the development of laminin-inspired hydrogels and the hurdles to overcome for these to progress from bench to bedside are discussed.
Collapse
Affiliation(s)
- Daniela Barros
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar , UPorto , Porto 4200-153 , Portugal
| | - Isabel F Amaral
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,FEUP - Faculdade de Engenharia , UPorto , Porto 4200-153 , Portugal
| | - Ana P Pêgo
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar , UPorto , Porto 4200-153 , Portugal.,FEUP - Faculdade de Engenharia , UPorto , Porto 4200-153 , Portugal
| |
Collapse
|
20
|
The Laminin- α1 Chain-Derived Peptide, AG73, Binds to Syndecans on MDA-231 Breast Cancer Cells and Alters Filopodium Formation. Anal Cell Pathol (Amst) 2019; 2019:9192516. [PMID: 31183318 PMCID: PMC6515157 DOI: 10.1155/2019/9192516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 02/17/2019] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is one of the most common forms of cancer affecting women in the United States, second only to skin cancers. Although treatments have been developed to combat primary breast cancer, metastasis remains a leading cause of death. An early step of metastasis is cancer cell invasion through the basement membrane. However, this process is not yet well understood. AG73, a synthetic laminin-α1 chain peptide, plays an important role in cell adhesion and has previously been linked to migration, invasion, and metastasis. Thus, we aimed to identify the binding partner of AG73 on breast cancer cells that could mediate cancer progression. We performed adhesion assays using MCF10A, T47D, SUM1315, and MDA-231 breast cell lines and found that AG73 binds to syndecans (Sdcs) 1, 2, and 4. This interaction was inhibited when we silenced Sdcs 1 and/or 4 in MDA-231 cells, indicating the importance of these receptors in this relationship. Through actin staining, we found that silencing of Sdc 1, 2, and 4 expression in MDA-231 cells exhibits a decrease in the length and number of filopodia bound to AG73. Expression of mouse Sdcs 1, 2, and 4 in MDA-231 cells provides rescue in filopodia, and overexpression of Sdcs 1 and 2 leads to increased filopodium length and number. Our findings demonstrate an intrinsic interaction between AG73 in the tumor environment and the Sdcs on breast cancer cells in supporting tumor cell adhesion and invasion through filopodia, an important step in cancer metastasis.
Collapse
|
21
|
Wilems T, Vardhan S, Wu S, Sakiyama-Elbert S. The influence of microenvironment and extracellular matrix molecules in driving neural stem cell fate within biomaterials. Brain Res Bull 2019; 148:25-33. [PMID: 30898579 DOI: 10.1016/j.brainresbull.2019.03.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/07/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
Transplantation of stem cells is a promising potential therapy for central nervous system disease and injury. The capacity for self-renewal, proliferation of progenitor cells, and multi-lineage potential underscores the need for controlling stem cell fate. Furthermore, transplantation within a hostile environment can lead to significant cell death and limited therapeutic potential. Tissue-engineered materials have been developed to both regulate stem cell fate, increase transplanted cell viability, and improve therapeutic outcomes. Traditionally, regulation of stem cell differentiation has been driven through soluble signals, such as growth factors. While these signals are important, insoluble factors from the local microenvironment or extracellular matrix (ECM) molecules also contribute to stem cell activity and fate. Understanding the microenvironment factors that influence stem cell fate, such as mechanical properties, topography, and presentation of specific ECM ligands, is necessary for designing improved biomaterials. Here we review some of the microenvironment factors that regulate stem cell fate and how they can be incorporated into biomaterials as part of potential CNS therapies.
Collapse
Affiliation(s)
- Thomas Wilems
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Sangamithra Vardhan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Siliang Wu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Shelly Sakiyama-Elbert
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, 78712, USA.
| |
Collapse
|
22
|
Follmann HD, Messias I, Queiroz MN, Araujo RA, Rubira AF, Silva R. Designing hybrid materials with multifunctional interfaces for wound dressing, electrocatalysis, and chemical separation. J Colloid Interface Sci 2019; 533:106-125. [DOI: 10.1016/j.jcis.2018.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023]
|
23
|
Sivaraman K, Shanthi C. Matrikines for therapeutic and biomedical applications. Life Sci 2018; 214:22-33. [PMID: 30449450 DOI: 10.1016/j.lfs.2018.10.056] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/25/2022]
Abstract
Matrikines, peptides originating from the fragmentation of extracellular matrix proteins are identified to play important role in both health and disease. They possess biological activities, much different from their parent protein. Identification of such bioactive cryptic regions in the extracellular matrix proteins has attracted the researchers all over the world in the recent decade. These bioactive peptides could find use in preparation of biomaterials and tissue engineering applications. Matrikines identified in major extracellular matrix (ECM) proteins like collagen, elastin, fibronectin, and laminin are being extensively studied for use in tissue engineering and regenerative medicine. They are identified to modulate cellular activity like cell growth, proliferation, migration and may induce apoptosis. RGD, a well-known peptide identified in fibronectin with cell adhesive property is being investigated in designing biomaterials. Collagen hexapeptide GFOGER was found to promote cell adhesion and differentiation. Laminin also possesses regions with strong cell adhesion property. Recently, cell-penetrating peptides from elastin are used as a targeted delivery system for therapeutic drugs. The continued search for cryptic sequences in the extracellular matrix proteins along with advanced peptide coupling chemistries would lead to biomaterials with improved surface properties. This review article outlines the peptides derived from extracellular matrix and some of the possible applications of these peptides in therapeutics and tissue engineering applications.
Collapse
Affiliation(s)
- K Sivaraman
- School of Biosciences and Technology, VIT, Vellore 632014, Tamilnadu, India
| | - C Shanthi
- School of Biosciences and Technology, VIT, Vellore 632014, Tamilnadu, India.
| |
Collapse
|
24
|
Fang YM, Lin DQ, Yao SJ. Review on biomimetic affinity chromatography with short peptide ligands and its application to protein purification. J Chromatogr A 2018; 1571:1-15. [DOI: 10.1016/j.chroma.2018.07.082] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/12/2018] [Accepted: 07/29/2018] [Indexed: 10/28/2022]
|
25
|
Kurokawa N, Hotta A. Thermomechanical properties of highly transparent self-reinforced polylactide composites with electrospun stereocomplex polylactide nanofibers. POLYMER 2018. [DOI: 10.1016/j.polymer.2018.08.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
26
|
Future Research Directions in the Design of Versatile Extracellular Matrix in Tissue Engineering. Int Neurourol J 2018; 22:S66-75. [PMID: 30068068 PMCID: PMC6077942 DOI: 10.5213/inj.1836154.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
Native and artificial extracellular matrices (ECMs) have been widely applied in biomedical fields as one of the most effective components in tissue regeneration. In particular, ECM-based drugs are expected to be applied to treat diseases in organs relevant to urology, because tissue regeneration is particularly important for preventing the recurrence of these diseases. Native ECMs provide a complex in vivo architecture and native physical and mechanical properties that support high biocompatibility. However, the applications of native ECMs are limited due to their tissue-specificity and chemical complexity. Artificial ECMs have been fabricated in an attempt to create a broadly applicable scaffold by using controllable components and a uniform formulation. On the other hands, artificial ECMs fail to mimic the properties of a native ECM; consequently, their applications in tissues are also limited. For that reason, the design of a versatile, hybrid ECM that can be universally applied to various tissues is an emerging area of interest in the biomedical field.
Collapse
|
27
|
Zarrintaj P, Manouchehri S, Ahmadi Z, Saeb MR, Urbanska AM, Kaplan DL, Mozafari M. Agarose-based biomaterials for tissue engineering. Carbohydr Polym 2018; 187:66-84. [PMID: 29486846 DOI: 10.1016/j.carbpol.2018.01.060] [Citation(s) in RCA: 346] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/28/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Abstract
Agarose is a natural polysaccharide polymer having unique characteristics that give reason to consider it for tissue engineering applications. Special characteristics of agarose such as its excellent biocompatibility, thermo-reversible gelation behavior and physiochemical features support its use as a biomaterial for cell growth and/or controlled/localized drug delivery. The resemblance of this natural carbohydrate polymer to the extracellular matrix results in attractive features that bring about a strong interest in its usage in the field. The scope of this review is to summarize the extensive researches addressing agarose-based biomaterials in order to provide an in-depth understanding of its tissue engineering-related applications.
Collapse
Affiliation(s)
- Payam Zarrintaj
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Saeed Manouchehri
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Zahed Ahmadi
- Department of Chemistry, Amirkabir University of Technology, Tehran, Iran
| | - Mohammad Reza Saeb
- Department of Resin and Additives, Institute for Color Science and Technology, P.O. Box: 16765-654, Tehran, Iran.
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
28
|
Chew KW, Juan JC, Phang SM, Ling TC, Show PL. An overview on the development of conventional and alternative extractive methods for the purification of agarose from seaweed. SEP SCI TECHNOL 2017. [DOI: 10.1080/01496395.2017.1394881] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kit Wayne Chew
- Nanotechnology & Catalysis Research Centre (NANOCAT), University of Malaya, Kuala Lumpur, Malaysia
- Department of Chemical and Environmental Engineering, Faculty of Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, Selangor Darul Ehsan, Malaysia
| | - Joon Ching Juan
- Nanotechnology & Catalysis Research Centre (NANOCAT), University of Malaya, Kuala Lumpur, Malaysia
- Laboratory of Advanced Catalysis and Environmental Technology, Monash University Sunway Campus, Bandar Sunway, Malaysia
| | - Siew Moi Phang
- Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
- Institute of Ocean and Earth Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Tau Chuan Ling
- Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Pau Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
29
|
Aso A, Taki K, Maeda T, Toma K, Tamiaki H, Hotta A. Composition‐dependent sol‐gel transition of amphiphilic blend of PEG with hydrophobic gallamide components. J Appl Polym Sci 2017. [DOI: 10.1002/app.45402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Akihiro Aso
- Department of Mechanical EngineeringKeio University3‐14‐1, Hiyoshi, Kohoku‐ku, Yokohama223‐8522 Japan
| | - Kazutaka Taki
- Graduate School of Life SciencesRitsumeikan UniversityNoji‐higashi 1‐1‐1, Kusatsu Shiga525‐8577 Japan
| | - Tomoki Maeda
- Department of Mechanical EngineeringKeio University3‐14‐1, Hiyoshi, Kohoku‐ku, Yokohama223‐8522 Japan
| | - Kazunori Toma
- Asahi Kasei Corporation2‐1, Samejima, Fuji Shizuoka416‐8501 Japan
| | - Hitoshi Tamiaki
- Graduate School of Life SciencesRitsumeikan UniversityNoji‐higashi 1‐1‐1, Kusatsu Shiga525‐8577 Japan
| | - Atsushi Hotta
- Department of Mechanical EngineeringKeio University3‐14‐1, Hiyoshi, Kohoku‐ku, Yokohama223‐8522 Japan
| |
Collapse
|
30
|
Regulation of human nucleus pulposus cells by peptide-coupled substrates. Acta Biomater 2017; 55:100-108. [PMID: 28433788 DOI: 10.1016/j.actbio.2017.04.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/20/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022]
Abstract
Nucleus pulposus (NP) cells are derived from the notochord and differ from neighboring cells of the intervertebral disc in phenotypic marker expression and morphology. Adult human NP cells lose this phenotype and morphology with age in a pattern that contributes to progressive disc degeneration and pathology. Select laminin-mimetic peptide ligands and substrate stiffnesses were examined for their ability to regulate human NP cell phenotype and biosynthesis through the expression of NP-specific markers aggrecan, N-cadherin, collagen types I and II, and GLUT1. Peptide-conjugated substrates demonstrated an ability to promote expression of healthy NP-specific markers, as well as increased biosynthetic activity. We show an ability to re-express markers of the juvenile NP cell and morphology through control of peptide presentation and stiffness on well-characterized polyacrylamide substrates. NP cells cultured on surfaces conjugated with α3 integrin receptor peptides P4 and P678, and on α2, α5, α6, β1 integrin-recognizing peptide AG10, show increased expression of aggrecan, N-cadherin, and types I and II collagen, suggesting a healthier, more juvenile-like phenotype. Multi-cell cluster formation was also observed to be more prominent on peptide-conjugated substrates. These findings indicate a critical role for cell-matrix interactions with specific ECM-mimetic peptides in supporting and maintaining a healthy NP cell phenotype and bioactivity. STATEMENT OF SIGNIFICANCE NP cells reside in a laminin-rich environment that deteriorates with age, including a loss of water content and changes in the extracellular matrix (ECM) structure that may lead to the development of a degenerated IVD. There is great interest in methods to re-express healthy, biosynthetically active NP cells using laminin-derived biomimetic peptides toward the goal of using autologous cell sources for tissue regeneration. Here, we describe a novel study utilizing several laminin mimetic peptides conjugated to polyacrylamide gels that are able to support an immature, healthy NP phenotype after culture on "soft" peptide gels. These findings can support future studies in tissue regeneration where cells may be directed to a desired regenerative phenotype using niche-specific ECM peptides.
Collapse
|
31
|
Miranda-Alarcón YS, Brown AM, Santora AM, Banerjee IA. Growth of Self-Assembled Bio-Organic Nanomatrices for Skin Tissue Engineering — An in vitro Study. ACTA ACUST UNITED AC 2016. [DOI: 10.1142/s1793984416500021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this paper, we have developed self-assembled nanoscale assemblies that were prepared by conjugating furan-2-carboxylic acid-3-aminopropyl amide with the short peptide sequence Gly-His (abbreviated Gly-His-FCAP). To mimic the extracellular matrix of mammalian fibroblasts and keratinocytes, the assemblies were then conjugated with Type I collagen. We then integrated the collagen bound Gly-His-FCAP assemblies with a short peptide sequence derived from salamander skin into the nanoscale assemblies for the first time to impart regenerative and wound healing properties to the composites. The antioxidant, antimicrobial and biodegradable properties were examined and results indicate that the nanocomposites displayed antioxidant properties as displayed by 1,1-diphenyl-2-picrylhydrazyl (DPPH) assay. The biodegradability was found to be gradual. The nanocomposites were also found to inhibit the growth of the fungus Rhizopus sporangia over an 18[Formula: see text]h growth period. As proof of concept, to demonstrate the development of three-dimensional (3D) engineered skin in vitro, 3D printed PLA scaffolds of 2.5[Formula: see text]mm thickness were submerged in media containing nanocomposites and co-cultures of dermal fibroblasts with epidermal keratinocytes mimicking three dimensional skin substitute was examined. Our results indicated that the nanocomposites adhered to and supported cell proliferation and mimicked the components of skin and may have potential applications in skin tissue regeneration.
Collapse
Affiliation(s)
| | - Alexandra M. Brown
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY 10458, USA
| | - Anthony M. Santora
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY 10458, USA
| | - Ipsita A. Banerjee
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, NY 10458, USA
| |
Collapse
|
32
|
Lilliu MA, Seo YJ, Isola M, Charbonneau AM, Zeitouni A, El-Hakim M, Tran SD. Natural extracellular matrix scaffolds recycled from human salivary digests: a morphometric study. Oral Dis 2016; 22:313-23. [PMID: 26785831 DOI: 10.1111/odi.12444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE A challenge in engineering tissues is to supply parenchymal cells with suitable scaffolds which ideally reproduce the extracellular matrix (ECM). This study tested the hypothesis of preserving the 'residual connective tissue' remaining after mechanical and enzymatic release of cells from human submandibular gland biopsies (that we named 'natural ExtraCellular Matrix scaffolds', nECMsc) to be used as recycled natural scaffolds. The objective was to test whether nECMsc and native salivary tissue were comparable morphologically, in ECM proteins composition, and in cell seeding efficiency. METHODS Following cell isolation procedures, nECMsc were kept, either fresh or frozen (sectioned into 12-μm-thick slices), and examined with high-resolution electron microscopy (HRSEM) for its three-dimensional structure, and with picrosirius red staining and immunogold staining for ECM protein composition and distribution, respectively. nECMsc were seeded with human epithelial cells and fibroblasts to assess cell attachment and proliferation in short-term experiments. RESULTS Under HRSEM, nECMsc had comparable fiber arrangement to original glands. Histochemical and immunogold-labeling examinations revealed the presence of collagen types I, III, and IV. Seeded epithelial cells and fibroblasts attached, proliferated (14-55%), and were alive (86-99%) after 4-8 days of culture. CONCLUSIONS nECMsc retained native ECM proteins and maintained their distribution. Seeded cells remained viable on nECMsc.
Collapse
Affiliation(s)
- M A Lilliu
- Faculty of Dentistry, McGill University, Montreal, QC, Canada.,Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Y J Seo
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - M Isola
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - A M Charbonneau
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - A Zeitouni
- Department of Otolaryngology, Hôpital Royale Victoria McGill University, Montreal, QC, Canada
| | - M El-Hakim
- Department of Oral and Maxillofacial Surgery, McGill University Health Centre, Montreal General Hospital McGill University, Montreal, QC, Canada
| | - S D Tran
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
33
|
Homogeneous tosylation of agarose as an approach toward novel functional polysaccharide materials. Carbohydr Polym 2015; 127:236-45. [DOI: 10.1016/j.carbpol.2015.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 11/19/2022]
|
34
|
Electrospinning of agar/PVA aqueous solutions and its relation with rheological properties. Carbohydr Polym 2015; 115:348-55. [DOI: 10.1016/j.carbpol.2014.08.074] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 08/08/2014] [Accepted: 08/10/2014] [Indexed: 11/22/2022]
|
35
|
Gómez-Mascaraque LG, Méndez JA, Fernández-Gutiérrez M, Vázquez B, San Román J. Oxidized dextrins as alternative crosslinking agents for polysaccharides: application to hydrogels of agarose-chitosan. Acta Biomater 2014; 10:798-811. [PMID: 24121253 DOI: 10.1016/j.actbio.2013.10.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/27/2013] [Accepted: 10/03/2013] [Indexed: 12/28/2022]
Abstract
Hydrogel networks that combine suitable physical and biomechanical characteristics for tissue engineering scaffolds are in demand. The aim of this work was the development of hydrogel networks based on agarose and chitosan using oxidized dextrins as low cytotoxicity crosslinking agents, paying special attention to the study of the influence of the polysaccharide composition and oxidation degree of the dextrins in the final characteristics of the network. The results show that the formation of an interpenetrating or a semi-interpenetrating polymer network was mainly dependent on a minimum agarose content and degree of oxidation of dextrin. Spectroscopic, thermal and swelling analysis revealed good compatibility with an absence of phase separation of polysaccharides at agarose:chitosan proportions of 50:50 and 25:75. The analysis of atomic force microscopy images showed the formation of a fibrillar microstructure whose distribution within the crosslinked chitosan depended mainly on the crosslinker. All materials exhibited the viscoelastic behaviour typical of gels, with a constant storage modulus independent of frequency for all compositions. The stiffness was strongly influenced by the degree of oxidation of the crosslinker. Cellular response to the hydrogels was studied with cells of different strains, and cell adhesion and proliferation was correlated with the homogeneity of the samples and their elastic properties. Some hydrogel formulations seemed to be candidates for tissue engineering applications such as wound healing or soft tissue regeneration.
Collapse
Affiliation(s)
- Laura G Gómez-Mascaraque
- CIBER-BBN, Ebro River Campus, R&D Building, Block 5, Floor 1, Poeta Mariano Esquillor s/n, 50017 Zaragoza, Spain; Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | - José Alberto Méndez
- Escola Politècnica Superior, Edifici PI, Campus Montilivi, University of Girona, 17071 Girona, Spain
| | - Mar Fernández-Gutiérrez
- CIBER-BBN, Ebro River Campus, R&D Building, Block 5, Floor 1, Poeta Mariano Esquillor s/n, 50017 Zaragoza, Spain; Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Blanca Vázquez
- CIBER-BBN, Ebro River Campus, R&D Building, Block 5, Floor 1, Poeta Mariano Esquillor s/n, 50017 Zaragoza, Spain; Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Julio San Román
- CIBER-BBN, Ebro River Campus, R&D Building, Block 5, Floor 1, Poeta Mariano Esquillor s/n, 50017 Zaragoza, Spain; Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| |
Collapse
|
36
|
Thiele J, Ma Y, Bruekers SMC, Ma S, Huck WTS. 25th anniversary article: Designer hydrogels for cell cultures: a materials selection guide. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:125-47. [PMID: 24227691 DOI: 10.1002/adma.201302958] [Citation(s) in RCA: 313] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/31/2013] [Indexed: 05/25/2023]
Abstract
Cell culturing, whether for tissue engineering or cell biology studies, always involves placing cells in a non-natural environment and no material currently exist that can mimic the entire complexity of natural tissues and variety of cell-matrix interactions that is found in vivo. Here, we review the vast range of hydrogels, composed of natural or synthetic polymers that provide a route to tailored microenvironments.
Collapse
Affiliation(s)
- Julian Thiele
- Institute for Molecules and Materials, Heyendaalseweg 135, 6525, AJ, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Otagiri D, Yamada Y, Hozumi K, Katagiri F, Kikkawa Y, Nomizu M. Cell attachment and spreading activity of mixed laminin peptide-chitosan membranes. Biopolymers 2013; 100:751-9. [DOI: 10.1002/bip.22303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 04/24/2013] [Accepted: 05/28/2013] [Indexed: 01/16/2023]
Affiliation(s)
- Dai Otagiri
- Department of Clinical Biochemistry; Faculty of Pharmacy, Tokyo University of Pharmacy and Life Sciences; Hachioji Tokyo 192-0392 Japan
| | - Yuji Yamada
- Department of Clinical Biochemistry; Faculty of Pharmacy, Tokyo University of Pharmacy and Life Sciences; Hachioji Tokyo 192-0392 Japan
| | - Kentaro Hozumi
- Department of Clinical Biochemistry; Faculty of Pharmacy, Tokyo University of Pharmacy and Life Sciences; Hachioji Tokyo 192-0392 Japan
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry; Faculty of Pharmacy, Tokyo University of Pharmacy and Life Sciences; Hachioji Tokyo 192-0392 Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry; Faculty of Pharmacy, Tokyo University of Pharmacy and Life Sciences; Hachioji Tokyo 192-0392 Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry; Faculty of Pharmacy, Tokyo University of Pharmacy and Life Sciences; Hachioji Tokyo 192-0392 Japan
| |
Collapse
|
38
|
Hyland LL, Taraban MB, Yu YB. Using Small-Angle Scattering Techniques to Understand Mechanical Properties of Biopolymer-Based Biomaterials. SOFT MATTER 2013; 9:10.1039/C3SM51209F. [PMID: 24273590 PMCID: PMC3835338 DOI: 10.1039/c3sm51209f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The design and engineering of innovative biopolymer-based biomaterials for a variety of biomedical applications should be based on the understanding of the relationship between their nanoscale structure and mechanical properties. Down the road, such understanding could be fundamental to tune the properties of engineered tissues, extracellular matrices for cell delivery and proliferation/differentiation, etc. In this tutorial review, we attempt to show in what way biomaterial structural data can help to understand the bulk material properties. We begin with some background on common types of biopolymers used in biomaterials research, discuss some typical mechanical testing techniques and then review how others in the field of biomaterials have utilized small-angle scattering for material characterization. Detailed examples are then used to show the full range of possible characterization techniques available for biopolymer-based biomaterials. Future developments in the area of material characterization by small-angle scattering will undoubtedly facilitate the use of structural data to control the kinetics of assembly and final properties of prospective biomaterials.
Collapse
Affiliation(s)
| | - Marc B. Taraban
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA. Fax: 301-315-9953; Tel: 301-405-2829
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA. Fax: 410-706-5017; Tel: 410-706-7514
| | - Y. Bruce Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA. Fax: 410-706-5017; Tel: 410-706-7514
| |
Collapse
|
39
|
Yamada Y, Hozumi K, Katagiri F, Kikkawa Y, Nomizu M. Laminin-111-derived peptide-hyaluronate hydrogels as a synthetic basement membrane. Biomaterials 2013; 34:6539-47. [DOI: 10.1016/j.biomaterials.2013.05.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/23/2013] [Indexed: 01/28/2023]
|
40
|
Kim M, Kim YH, Tae G. Human mesenchymal stem cell culture on heparin-based hydrogels and the modulation of interactions by gel elasticity and heparin amount. Acta Biomater 2013; 9:7833-44. [PMID: 23643605 DOI: 10.1016/j.actbio.2013.04.041] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/01/2013] [Accepted: 04/24/2013] [Indexed: 11/25/2022]
Abstract
Human adipose-derived stem cells (hADSCs) are a promising cell source for tissue engineering and regenerative medicine with no ethnical issue and easy access of large quantities. Conventional surfaces for hADSC culture, such as tissue culture plates (TCPs), do not provide optimal environmental cues, leading to limited expansion, loss of pluripotency and undesirable differentiation of stem cells. The present study demonstrated that heparin-based hydrogels without additional modification provided an excellent surface for adhesion and proliferation of hADSCs, which were further tunable by both the amount of heparin (in a positive way) and the elasticity of hydrogel (in a negative way). The optimized heparin-based hydrogel could selectively modulate the adhesion of hADSCs and human bone marrow stem cells (but not all kinds of cells), and resulted in a significant increase in cell proliferation compared to TCP. Furthermore, in terms of the maintenance of pluripotency and specific differentiation, heparin-based hydrogel was much superior to TCP. The selective binding and proliferation of human mesenchymal stem cells on heparin-based hydrogel over other hydrogels were largely mediated by integrin β1 and selectin, and these superior characteristics were observed regardless of the presence of serum proteins in the culture medium. Consequently, heparin-based hydrogel could be a powerful platform for cultivation of mesenchymal stem cells in various applications.
Collapse
|
41
|
Pradhan-Bhatt S, Harrington DA, Duncan RL, Jia X, Witt RL, Farach-Carson MC. Implantable three-dimensional salivary spheroid assemblies demonstrate fluid and protein secretory responses to neurotransmitters. Tissue Eng Part A 2013; 19:1610-20. [PMID: 23442148 DOI: 10.1089/ten.tea.2012.0301] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Radiation treatment in patients with head and neck tumors commonly results in hyposalivation and xerostomia due to the loss of fluid-secreting salivary acinar cells. Patients develop susceptibility to oral infections, dental caries, impaired speech and swallowing, reducing the quality of life. Clinical management is largely unsatisfactory. The development of a tissue-engineered, implantable salivary gland will greatly benefit patients suffering from xerostomia. This report compares the ability of a 2.5-dimensional (2.5D) and a three-dimensional (3D) hyaluronic acid (HA)-based culture system to support functional salivary units capable of producing fluid and phenotypic proteins. Parotid cells seeded on 2.5D, as well as those encapsulated in 3D HA hydrogels, self-assembled into acini-like structures and expressed functional neurotransmitter receptors. Structures in 3D hydrogels merged to form organized 50 μm spheroids that could be maintained in culture for over 100 days and merged to form structures over 500 μm in size. Treatment of acini-like structures with the β-adrenergic agonists norepinephrine or isoproterenol increased granule production and α-amylase staining in treated structures, demonstrating regain of protein secretion. Upon treatment with the M3 muscarinic agonist acetylcholine, acini-like structures activated the fluid production pathway by increasing intracellular calcium levels. The increase in intracellular calcium seen in structures in the 3D hydrogel culture system was more robust and prolonged than that in 2.5D. To compare the long-term survival and retention of acini-like structures in vivo, cell-seeded 2.5D and 3D hydrogels were implanted into an athymic rat model. Cells in 2.5D failed to maintain organized acini-like structures and dispersed in the surrounding tissue. Encapsulated cells in 3D retained their spheroid structure and structural integrity, along with the salivary biomarkers and maintained viability for over 3 weeks in vivo. This report identifies a novel hydrogel culture system capable of creating and maintaining functional 3D salivary spheroid structures for long periods in vitro that regain both fluid and protein secreting functions and are suitable for tissue restoration.
Collapse
Affiliation(s)
- Swati Pradhan-Bhatt
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | | | | | | | | | | |
Collapse
|
42
|
Zhao W, Jin X, Cong Y, Liu Y, Fu J. Degradable natural polymer hydrogels for articular cartilage tissue engineering. JOURNAL OF CHEMICAL TECHNOLOGY & BIOTECHNOLOGY 2013; 88:327-339. [DOI: 10.1002/jctb.3970] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 09/27/2012] [Indexed: 01/04/2025]
Abstract
AbstractArticular cartilage has poor ability to heal once damaged. Tissue engineering with scaffolds of polymer hydrogels is promising for cartilage regeneration and repair. Polymer hydrogels composed of highly hydrated crosslinked networks mimic the collagen networks of the cartilage extracellular matrix and thus are employed as inserts at cartilage defects not only to temporarily relieve the pain but also to support chondrocyte proliferation and neocartilage regeneration. The biocompatibility, biofunctionality, mechanical properties, and degradation of the polymer hydrogels are the most important parameters for hydrogel‐based cartilage tissue engineering. Degradable biopolymers with natural origin have been widely used as biomaterials for tissue engineering because of their outstanding biocompatibility, low immunological response, low cytotoxicity, and excellent capability to promote cell adhesion, proliferation, and regeneration of new tissues. This review covers several important natural proteins (collagen, gelatin, fibroin, and fibrin) and polysaccharides (chitosan, hyaluronan, alginate and agarose) widely used as hydrogels for articular cartilage tissue engineering. The mechanical properties, structures, modification, and structure–performance relationship of these hydrogels are discussed since the chemical structures and physical properties dictate the in vivo performance and applications of polymer hydrogels for articular cartilage regeneration and repair. © 2012 Society of Chemical Industry
Collapse
Affiliation(s)
- Wen Zhao
- Department of Orthopedic Surgery Beijing Aero‐space General Hospital Beijing China
| | - Xing Jin
- Clemson‐MUSC Bioengineering Joint Program Charleston SC 29425 USA
| | - Yang Cong
- School of Chemical Engineering Ningbo University of Technology Ningbo 315016 China
| | - Yuying Liu
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics Medical University of South Carolina Charleston SC 29425 USA
| | - Jun Fu
- Ningbo Key Laboratory of Polymer Materials, Polymers and Composites Division, Ningbo Institute of Materials Technology and Engineering Chinese Academy of Sciences Ningbo Zhejiang 315201 China
| |
Collapse
|
43
|
Kikkawa Y, Hozumi K, Katagiri F, Nomizu M, Kleinman HK, Koblinski JE. Laminin-111-derived peptides and cancer. Cell Adh Migr 2012; 7:150-256. [PMID: 23263633 PMCID: PMC3544779 DOI: 10.4161/cam.22827] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Laminin-111 is a large trimeric basement membrane glycoprotein with many active sites. In particular, four peptides active in tumor malignancy studies have been identified in laminin-111 using a systematic peptide screening method followed by various assays. Two of the peptides (IKVAV and AG73) are found on the α1 chain, one (YIGSR) of the β1 chain and one (C16) on the γ1 chain. The four peptides have distinct activities and receptors. Since three of the peptides (IKVAV, AG73 and C16) strongly promote tumor growth, this may explain the potent effects laminin-111 has on malignant cells. The peptide, YIGSR, decreases tumor growth and experimental metastasis via a 32/67 kD receptor while IKVAV increases tumor growth, angiogenesis and protease activity via integrin receptors. AG73 increases tumor growth and metastases via syndecan receptors. C16 increases tumor growth and angiogenesis via integrins. Identification of such sites on laminin-111 will have use in defining strategies to develop therapeutics for cancer.
Collapse
Affiliation(s)
- Yamato Kikkawa
- Laboratory of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|