1
|
Zeng L, Zeng J, He J, Zhou Y, Li Y, Li C, Lin Z, Chen G, Wu H, Zhou L. ALB-PRF facilitates chondrogenesis by promoting chondrocytes migration, proliferation and differentiation. Platelets 2024; 35:2414792. [PMID: 39431681 DOI: 10.1080/09537104.2024.2414792] [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: 05/16/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
Cartilage injury is common in orthopedics and cartilage tissue engineering provides a therapeutic direction for cartilage regeneration. Albumin (ALB)-platelet-rich fibrin (PRF) is speculated to be an ideal natural scaffold material for cartilage tissue engineering theoretically as a product derived from human venous blood. Through in vitro and in vivo experiments, it was demonstrated that ALB-PRF displayed porous structure and slowly released growth factors (TGF-β1, PDGF-AA, PDGF-AB, PDGF-BB, EGF, IGF-1 and VEGF), ALB-PRF conditioned media promoted proliferation, migration, adhesion, phenotype maintenance and extracellular matrix secretion of rabbit chondrocytes. Moreover, ALB-PRF facilitated chondrogenesis in vivo, the regenerative cartilage formed by ALB-PRF/chondrocytes was histologically similar to that of natural knee joint cartilage, the regenerative cartilage expressed cartilage differentiation marker (SOX9, ACAN and COL II), and proliferation marker PCNA and secreted abundant glycosaminoglycans (GAGs) in extracellular matrix. In conclusion, ALB-PRF promoted the migration, proliferation and phenotype maintenance of chondrocytes in vitro. Its loose, porous structure and rich growth factors contained enhanced cell adhesion and growing into the materials. ALB-PRF facilitated chondrogenesis of chondrocytes in vivo.
Collapse
Affiliation(s)
- Lijuan Zeng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jun Zeng
- Department of General Dentistry and Oral Emergency, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jianfeng He
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yang Zhou
- Department of Oral and Maxillofacial Surgery, Dongfeng Stomatological Hospital, Hubei University of Medicine, Shiyan, China
| | - Yongqi Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Chengwei Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Zhiyan Lin
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Guangwei Chen
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Huilin Wu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Libin Zhou
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Torabi Rahvar P, Abdekhodaie MJ, Jooybar E, Gantenbein B. An enzymatically crosslinked collagen type II/hyaluronic acid hybrid hydrogel: A biomimetic cell delivery system for cartilage tissue engineering. Int J Biol Macromol 2024; 279:134614. [PMID: 39127277 DOI: 10.1016/j.ijbiomac.2024.134614] [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: 12/27/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
This study presents new injectable hydrogels based on hyaluronic acid and collagen type II that mimic the polysaccharide-protein structure of natural cartilage. After collagen isolation from chicken sternal cartilage, tyramine-grafted hyaluronic acid and collagen type II (HA-Tyr and COL-II-Tyr) were synthesized. Hybrid hydrogels were prepared with different ratios of HA-Tyr/COL-II-Tyr using horseradish peroxidase and noncytotoxic concentrations of hydrogen peroxide to encapsulate human bone marrow-derived mesenchymal stromal cells (hBM-MSCs). The findings showed that a higher HA-Tyr content resulted in a higher storage modulus and a lower hydrogel shrinkage, resulting in hydrogel swelling. Incorporating COL-II-Tyr into HA-Tyr hydrogels induced a more favorable microenvironment for hBM-MSCs chondrogenic differentiation. Compared to HA-Tyr alone, the hybrid HA-Tyr/COL-II-Tyr hydrogel promoted enhanced chondrocyte adhesion, spreading, proliferation, and upregulation of cartilage-related gene expression. These results highlight the promising potential of injectable HA-Tyr/COL-II-Tyr hybrid hydrogels to deliver cells for cartilage regeneration.
Collapse
Affiliation(s)
- Parisa Torabi Rahvar
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Mohammad J Abdekhodaie
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Environmental and Applied Science Management, Yeates School of Graduate Studies, Toronto Metropolitan University, Toronto, Canada.
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland; Inselspital, Bern University Hospital, Department of Orthopedic Surgery & Traumatology, Bern, Switzerland
| |
Collapse
|
3
|
Babiak PM, Battistoni CM, Cahya L, Athreya R, Minnich J, Panitch A, Liu JC. Tunable Blended Collagen I/II and Collagen I/III Hydrogels as Tissue Mimics. Macromol Biosci 2024:e2400280. [PMID: 39427345 DOI: 10.1002/mabi.202400280] [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: 06/13/2024] [Revised: 10/03/2024] [Indexed: 10/22/2024]
Abstract
Collagen (Col) is commonly used as a natural biomaterial for biomedical applications. Although Col I is the most prevalent col type employed, many collagen types work together in vivo to confer function and biological activity. Thus, blending collagen types can better recapitulate many native environments. This work investigates how hydrogel properties can be tuned through blending collagen types (col I/II and col I/III) and by varying polymerization temperatures. Col I/II results in poorly developed fibril networks, which softened the gels, especially at lower polymerization temperatures. Conversely, col I/III hydrogels exhibit well-connected fibril networks with localized areas of fine fibrils and result in stiffer hydrogels. A decreased molecular mass recovery rate is observed in blended hydrogels. The altered fibril morphologies, mechanical properties, and biological signals of the blended gels can be leveraged to alter cell responses and can be used as models for different tissue types (e.g., healthy vs fibrotic tissue). Furthermore, the biomimetic hydrogel properties are a tool that can be used to modulate the transport of drugs, nutrients, and wastes in tissue engineering applications.
Collapse
Affiliation(s)
- Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Carly M Battistoni
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Leonard Cahya
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Rithika Athreya
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Jason Minnich
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Alyssa Panitch
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
4
|
Yu L, Wu H, Zeng S, Hu X, Wu Y, Zhou J, Yuan L, Zhang Q, Xiang C, Feng Z. Menstrual blood-derived mesenchymal stem cells combined with collagen I gel as a regenerative therapeutic strategy for degenerated disc after discectomy in rats. Stem Cell Res Ther 2024; 15:75. [PMID: 38475906 DOI: 10.1186/s13287-024-03680-w] [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: 06/10/2023] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Annulus fibrosis (AF) defects have been identified as the primary cause of disc herniation relapse and subsequent disc degeneration following discectomy. Stem cell-based tissue engineering offers a promising approach for structural repair. Menstrual blood-derived mesenchymal stem cells (MenSCs), a type of adult stem cell, have gained attention as an appealing source for clinical applications due to their potential for structure regeneration, with ease of acquisition and regardless of ethical issues. METHODS The differential potential of MenSCs cocultured with AF cells was examined by the expression of collagen I, SCX, and CD146 using immunofluorescence. Western blot and ELISA were used to examine the expression of TGF-β and IGF-I in coculture system. An AF defect animal model was established in tail disc of Sprague-Dawley rats (males, 8 weeks old). An injectable gel containing MenSCs (about 1*106/ml) was fabricated and transplanted into the AF defects immediately after the animal model establishment, to evaluate its repairment properties. Disc degeneration was assessed via magnetic resonance (MR) imaging and histological staining. Immunohistochemical analysis was performed to assess the expression of aggrecan, MMP13, TGF-β and IGF-I in discs with different treatments. Apoptosis in the discs was evaluated using TUNEL, caspase3, and caspase 8 immunofluorescence staining. RESULTS Coculturing MenSCs with AF cells demonstrated ability to express collagen I and biomarkers of AF cells. Moreover, the coculture system presented upregulation of the growth factors TGF-β and IGF-I. After 12 weeks, discs treated with MenSCs gel exhibited significantly lower Pffirrmann scores (2.29 ± 0.18), compared to discs treated with MenSCs (3.43 ± 0.37, p < 0.05) or gel (3.71 ± 0.29, p < 0.01) alone. There is significant higher MR index in disc treated with MenSCs gel than that treated with MenSCs (0.51 ± 0.05 vs. 0.24 ± 0.04, p < 0.01) or gel (0.51 ± 0.05 vs. 0.26 ± 0.06, p < 0.01) alone. Additionally, MenSCs gel demonstrated preservation of the structure of degenerated discs, as indicated by histological scoring (5.43 ± 0.43 vs. 9.71 ± 1.04 in MenSCs group and 10.86 ± 0.63 in gel group, both p < 0.01), increased aggrecan expression, and decreased MMP13 expression in vivo. Furthermore, the percentage of TUNEL and caspase 3-positive cells in the disc treated with MenSCs Gel was significantly lower than those treated with gel alone and MenSCs alone. The expression of TGF-β and IGF-I was higher in discs treated with MenSCs gel or MenSCs alone than in those treated with gel alone. CONCLUSION MenSCs embedded in collagen I gel has the potential to preserve the disc structure and prevent disc degeneration after discectomy, which was probably attributed to the paracrine of growth factors of MenSCs.
Collapse
Affiliation(s)
- Li Yu
- Department of Operating room, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Honghao Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shumei Zeng
- Department of gynaecology, Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojian Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuxu Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinhong Zhou
- Department of gynaecology, Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Yuan
- Innovative Precision Medicine (IPM) Group, Hangzhou, Zhejiang, China
| | - Qingqing Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhiyun Feng
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- , Building 8-2, 58#, Chengzhan Road, Hangzhou, 310003, China.
| |
Collapse
|
5
|
Nashchekina Y, Guryanov E, Lihachev A, Vaganov G, Popova E, Mikhailova N, Nashchekin A. Effect of Phytic Acid Addition on the Structure of Collagen-Hyaluronic Acid Composite Gel. Gels 2023; 9:963. [PMID: 38131949 PMCID: PMC10743047 DOI: 10.3390/gels9120963] [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: 11/16/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Composite collagen gels with hyaluronic acid are developed tissue-engineered structures for filling and regeneration of defects in various organs and tissues. For the first time, phytic acid was used to increase the stability and improve the mechanical properties of collagen gels with hyaluronic acid. Phytic acid is a promising cross-linker for collagen hydrogels and is a plant-derived antioxidant found in rich sources of beans, grains, and oilseeds. Phytic acid has several benefits due to its antioxidant, anticancer, and antitumor properties. In this work, studies were carried out on the kinetics of the self-assembly of collagen molecules in the presence of phytic and hyaluronic acids. It was shown that both of these acids do not lead to collagen self-assembly. Scanning electron microscopy showed that in the presence of phytic and hyaluronic acids, the collagen fibrils had a native structure, and the FTIR method confirmed the chemical cross-links between the collagen fibrils. DSC and rheological studies demonstrated that adding the phytic acid improved the stability and modulus of elasticity of the collagen gel. The presence of hyaluronic acid in the collagen gel slightly reduced the effect of phytic acid. The presence of phytic acid in the collagen gel improved the stability of the scaffold, but, after 1 week of cultivation, slightly reduced the viability of mesenchymal stromal cells cultured in the gel. The collagen type I gel with hyaluronic and phytic acids can be used to replace tissue defects, especially after the removal of cancerous tumors.
Collapse
Affiliation(s)
- Yuliya Nashchekina
- Center of Cell Technologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Pr. 4, 194064 St. Petersburg, Russia; (E.G.); (N.M.)
| | - Evgeny Guryanov
- Center of Cell Technologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Pr. 4, 194064 St. Petersburg, Russia; (E.G.); (N.M.)
| | - Alexey Lihachev
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya St. 26, 194021 St. Petersburg, Russia; (A.L.); (A.N.)
| | - Gleb Vaganov
- Institute of Macromolecular Compounds of Russian Academy of Sciences, V.O., Bol’shoy Pr. 31, 199004 St. Petersburg, Russia; (G.V.); (E.P.)
| | - Elena Popova
- Institute of Macromolecular Compounds of Russian Academy of Sciences, V.O., Bol’shoy Pr. 31, 199004 St. Petersburg, Russia; (G.V.); (E.P.)
| | - Natalya Mikhailova
- Center of Cell Technologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Pr. 4, 194064 St. Petersburg, Russia; (E.G.); (N.M.)
| | - Alexey Nashchekin
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya St. 26, 194021 St. Petersburg, Russia; (A.L.); (A.N.)
| |
Collapse
|
6
|
Torres JE, Meng F, Bhattacharya S, Buno KP, Ahmadzadegan A, Madduri S, Babiak PM, Vlachos PP, Solorio L, Yeo Y, Liu JC. Interpenetrating Networks of Collagen and Hyaluronic Acid That Serve as In Vitro Tissue Models for Assessing Macromolecular Transport. Biomacromolecules 2023; 24:4718-4730. [PMID: 37651737 DOI: 10.1021/acs.biomac.3c00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
High-fidelity preclinical in vitro tissue models can reduce the failure rate of drugs entering clinical trials. Collagen and hyaluronic acid (HA) are major components of the extracellular matrix of many native tissues and affect therapeutic macromolecule diffusion and recovery through tissues. Although collagen and HA are commonly used in tissue engineering, the physical and mechanical properties of these materials are variable and depend highly on processing conditions. In this study, HA was chemically modified and crosslinked via hydrazone bonds to form interpenetrating networks of crosslinked HA (HAX) with collagen (Col). These networks enabled a wide range of mechanical properties, including stiffness and swellability, and microstructures, such as pore morphology and size, that can better recapitulate diverse tissues. We utilized these interpenetrating ColHAX hydrogels as in vitro tissue models to examine macromolecular transport and recovery for early-stage drug screening. Hydrogel formulations with varying collagen and HAX concentrations imparted different gel properties based on the ratio of collagen to HAX. These gels were stable and swelled up to 170% of their original mass, and the storage moduli of the ColHAX gels increased over an order of magnitude by increasing collagen and HA concentration. Interestingly, when HAX concentration was constant and collagen concentration increased, both the pore size and spatial colocalization of collagen and HA increased. HA in the system dominated the ζ-potentials of the gels. The hydrogel and macromolecule properties impacted the mass transport and recovery of lysozyme, β-lactoglobulin, and bovine serum albumin (BSA) from the ColHAX gels─large molecules were largely impacted by mesh size, whereas small molecules were influenced primarily by electrostatic forces. Overall, the tunable properties demonstrated by the ColHAX hydrogels can be used to mimic different tissues for early-stage assays to understand drug transport and its relationship to matrix properties.
Collapse
Affiliation(s)
- Jessica E Torres
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Fanfei Meng
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sayantan Bhattacharya
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kevin P Buno
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Adib Ahmadzadegan
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sathvik Madduri
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pavlos P Vlachos
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
7
|
Nair A, Lin CY, Hsu FC, Wong TH, Chuang SC, Lin YS, Chen CH, Campagnola P, Lien CH, Chen SJ. Categorization of collagen type I and II blend hydrogel using multipolarization SHG imaging with ResNet regression. Sci Rep 2023; 13:19534. [PMID: 37945626 PMCID: PMC10636134 DOI: 10.1038/s41598-023-46417-0] [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: 09/05/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Previously, the discrimination of collagen types I and II was successfully achieved using peptide pitch angle and anisotropic parameter methods. However, these methods require fitting polarization second harmonic generation (SHG) pixel-wise information into generic mathematical models, revealing inconsistencies in categorizing collagen type I and II blend hydrogels. In this study, a ResNet approach based on multipolarization SHG imaging is proposed for the categorization and regression of collagen type I and II blend hydrogels at 0%, 25%, 50%, 75%, and 100% type II, without the need for prior time-consuming model fitting. A ResNet model, pretrained on 18 progressive polarization SHG images at 10° intervals for each percentage, categorizes the five blended collagen hydrogels with a mean absolute error (MAE) of 0.021, while the model pretrained on nonpolarization images exhibited 0.083 MAE. Moreover, the pretrained models can also generally regress the blend hydrogels at 20%, 40%, 60%, and 80% type II. In conclusion, the multipolarization SHG image-based ResNet analysis demonstrates the potential for an automated approach using deep learning to extract valuable information from the collagen matrix.
Collapse
Affiliation(s)
- Anupama Nair
- College of Photonics, National Yang Ming Chiao Tung University, Tainan, Taiwan
| | - Chun-Yu Lin
- College of Photonics, National Yang Ming Chiao Tung University, Tainan, Taiwan
| | - Feng-Chun Hsu
- College of Photonics, National Yang Ming Chiao Tung University, Tainan, Taiwan
| | - Ta-Hsiang Wong
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Paul Campagnola
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Chi-Hsiang Lien
- Department of Mechanical Engineering, National United University, Miaoli, Taiwan.
| | - Shean-Jen Chen
- College of Photonics, National Yang Ming Chiao Tung University, Tainan, Taiwan.
| |
Collapse
|
8
|
Do NT, Lee SY, Lee YS, Shin C, Kim D, Lee TG, Son JG, Kim SH. Time-sequential fibroblast-to-myofibroblast transition in elastin-variable 3D hydrogel environments by collagen networks. Biomater Res 2023; 27:103. [PMID: 37848974 PMCID: PMC10583321 DOI: 10.1186/s40824-023-00439-x] [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: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Fibrosis plays an important role in both normal physiological and pathological phenomena as fibroblasts differentiate to myofibroblasts. The activation of fibroblasts is determined through interactions with the surrounding extracellular matrix (ECM). However, how this fibroblast-to-myofibroblast transition (FMT) is regulated and affected by elastin concentration in a three-dimensional (3D) microenvironment has not been investigated. METHODS We developed an insoluble elastin-gradient 3D hydrogel system for long-lasting cell culture and studied the molecular mechanisms of the FMT in embedded cells by nanoflow LC-MS/MS analysis along with validation through real-time PCR and immunofluorescence staining. RESULTS By optimizing pH and temperature, four 3D hydrogels containing fibroblasts were successfully fabricated having elastin concentrations of 0, 20, 50, and 80% in collagen. At the low elastin level (20%), fibroblast proliferation was significantly increased compared to others, and in particular, the FMT was clearly observed in this condition. Moreover, through mass spectrometry of the hydrogel environment, it was confirmed that differentiation proceeded in two stages. In the early stage, calcium-dependent proteins including calmodulin and S100A4 were highly associated. On the other hand, in the late stage after several passages of cells, distinct markers of myofibroblasts were presented such as morphological changes, increased production of ECM, and increased α-SMA expression. We also demonstrated that the low level of elastin concentration induced some cancer-associated fibroblast (CAF) markers, including PDGFR-β, and fibrosis-related disease markers, including THY-1. CONCLUSION Using our developed 3D elastin-gradient hydrogel system, we evaluated the effect of different elastin concentrations on the FMT. The FMT was induced even at a low concentration of elastin with increasing CAF level via calcium signaling. With this system, we were able to analyze varying protein expressions in the overall FMT process over several cellular passages. Our results suggest that the elastin-gradient system employing nonlinear optics imaging provides a good platform to study activated fibroblasts interacting with the microenvironment, where the ECM plays a pivotal role.
Collapse
Affiliation(s)
- Nhuan T Do
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- BioMedical Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Sun Young Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Yoon Seo Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - ChaeHo Shin
- Interdisciplinary Materials Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- Nanoconvergence Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Daeho Kim
- Bruker Nano Surface & Metrology, Bruker Korea, Seongnam, 13493, Republic of Korea
| | - Tae Geol Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- Nanoconvergence Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Jin Gyeong Son
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
| | - Se-Hwa Kim
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
- BioMedical Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
9
|
Dartora VFC, Passos JS, Osorio B, Hung RC, Nguyen M, Wang A, Panitch A. Chitosan hydrogels with MK2 inhibitor peptide-loaded nanoparticles to treat atopic dermatitis. J Control Release 2023; 362:591-605. [PMID: 37660990 DOI: 10.1016/j.jconrel.2023.08.061] [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: 12/06/2022] [Revised: 08/05/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder that lacks ideal long-term treatment options due to a series of side effects, such as skin atrophy, related to the most common treatment prescribed to manage moderate-to-severe AD. In this study, a cell-penetrating MK2 inhibitor peptide YARA (YARAAARQARAKALNRQGLVAA) was loaded into hollow thermo-responsive pNIPAM nanoparticles (NP), which were further incorporated into chitosan hydrogels (H-NP-YARA) to promote local drug delivery, improve moisture and the anti-inflammatory activity. The NPs exhibited high loading efficiency (>50%) and the hydrogel remained porous following NP incorporation as observed by scanning electron microscopy (SEM). Both nanoparticles and hydrogels were able to improve the release of YARA and sustained release to up to 120 h. The hydrogels and NPs delivered 2 and 4-fold more YARA into viable skin layers of porcine skin in vitro at 12 h post-application than the non-encapsulated compound in intact and impaired barrier conditions. Furthermore, the YARA-loaded NPs (NP-YARA) and H-NP-YARA treatment decreased the levels of inflammatory cytokines up to 20 time-fold compared with the non-treated group of human keratinocytes under inflammatory conditions. Consistent with the results in cell culture, the loading of YARA in NP reduced the levels of IL-1β, IL-6, and TNF-α up to 3.3 times in an ex vivo skin culture model after induction of inflammation. A further decrease of up to 17 times-fold was observed with H-NP-YARA treatment compared to the drug in solution. Our data collectively suggest that chitosan hydrogel containing YARA-loaded nanoparticles is a promising new formulation for the topical treatment of AD.
Collapse
Affiliation(s)
- Vanessa F C Dartora
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Julia Sapienza Passos
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Institute of Biomedical Sciences, Department of Pharmacology, University of Sao Paulo, Brazil
| | - Blanca Osorio
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Ruei-Chun Hung
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Michael Nguyen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Aijun Wang
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Alyssa Panitch
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
10
|
Vinod A, Tadmor R, Katoshevski D, Gutmark EJ. Gels That Serve as Mucus Simulants: A Review. Gels 2023; 9:555. [PMID: 37504435 PMCID: PMC10379079 DOI: 10.3390/gels9070555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/29/2023] Open
Abstract
Mucus is a critical part of the human body's immune system that traps and carries away various particulates such as anthropogenic pollutants, pollen, viruses, etc. Various synthetic hydrogels have been developed to mimic mucus, using different polymers as their backbones. Common to these simulants is a three-dimensional gel network that is physically crosslinked and is capable of loosely entrapping water within. Two of the challenges in mimicking mucus using synthetic hydrogels include the need to mimic the rheological properties of the mucus and its ability to capture particulates (its adhesion mechanism). In this paper, we review the existing mucus simulants and discuss their rheological, adhesive, and tribological properties. We show that most, but not all, simulants indeed mimic the rheological properties of the mucus; like mucus, most hydrogel mucus simulants reviewed here demonstrated a higher storage modulus than its loss modulus, and their values are in the range of that found in mucus. However, only one mimics the adhesive properties of the mucus (which are critical for the ability of mucus to capture particulates), Polyvinyl alcohol-Borax hydrogel.
Collapse
Affiliation(s)
- Appu Vinod
- Department of Mechanical Engineering, Ben Gurion University, Beer Sheva 84105, Israel
| | - Rafael Tadmor
- Department of Mechanical Engineering, Ben Gurion University, Beer Sheva 84105, Israel
| | - David Katoshevski
- Department of Civil and Environmental Engineering, Ben Gurion University, Beer Sheva 84105, Israel
| | - Ephraim J Gutmark
- Department of Aerospace Engineering & Engineering Mechanics, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
11
|
Kilian D, Poddar A, Desrochers V, Heinemann C, Halfter N, Liu S, Rother S, Gelinsky M, Hintze V, Lode A. Cellular adhesion and chondrogenic differentiation inside an alginate-based bioink in response to tailorable artificial matrices and tannic acid treatment. BIOMATERIALS ADVANCES 2023; 147:213319. [PMID: 36758282 DOI: 10.1016/j.bioadv.2023.213319] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/30/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Many established bioinks fulfill important requirements regarding fabrication standards and cytocompatibility. Current research focuses on development of functionalized bioinks with an improved support of tissue-specific cell differentiation. Many approaches primarily depend on decellularized extracellular matrices or blood components. In this study, we investigated the combination of a highly viscous alginate-methylcellulose (algMC) bioink with collagen-based artificial extracellular matrix (aECM) as a finely controllable and tailorable system composed of collagen type I (col) with and without chondroitin sulfate (CS) or sulfated hyaluronan (sHA). As an additional stabilizer, the polyphenol tannic acid (TA) was integrated into the inks. The assessment of rheological properties and printability as well as hydrogel microstructure revealed no adverse effect of the integrated components on the inks. Viability, adhesion, and proliferation of bioprinted immortalized human mesenchymal stem cells (hTERT-MSC) was improved indicating enhanced interaction with the designed microenvironment. Furthermore, chondrogenic matrix production (collagen type II and sulfated glycosaminoglycans) by primary human chondrocytes (hChon) was enhanced by aECM. Supplementing the inks with TA was required for these positive effects but caused cytotoxicity as soon as TA concentrations exceeded a certain amount. Thus, combining tailorable aECM with algMC and balanced TA addition proved to be a promising approach for promoting adhesion of immortalized stem cells and differentiation of chondrocytes in bioprinted scaffolds.
Collapse
Affiliation(s)
- David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Aayush Poddar
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Vanessa Desrochers
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Christiane Heinemann
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Norbert Halfter
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Suihong Liu
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany; Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany; Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Saar, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
12
|
McCourt JL, Stearns-Reider KM, Mamsa H, Kannan P, Afsharinia MH, Shu C, Gibbs EM, Shin KM, Kurmangaliyev YZ, Schmitt LR, Hansen KC, Crosbie RH. Multi-omics analysis of sarcospan overexpression in mdx skeletal muscle reveals compensatory remodeling of cytoskeleton-matrix interactions that promote mechanotransduction pathways. Skelet Muscle 2023; 13:1. [PMID: 36609344 PMCID: PMC9817407 DOI: 10.1186/s13395-022-00311-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The dystrophin-glycoprotein complex (DGC) is a critical adhesion complex of the muscle cell membrane, providing a mechanical link between the extracellular matrix (ECM) and the cortical cytoskeleton that stabilizes the sarcolemma during repeated muscle contractions. One integral component of the DGC is the transmembrane protein, sarcospan (SSPN). Overexpression of SSPN in the skeletal muscle of mdx mice (murine model of DMD) restores muscle fiber attachment to the ECM in part through an associated increase in utrophin and integrin adhesion complexes at the cell membrane, protecting the muscle from contraction-induced injury. In this study, we utilized transcriptomic and ECM protein-optimized proteomics data sets from wild-type, mdx, and mdx transgenic (mdxTG) skeletal muscle tissues to identify pathways and proteins driving the compensatory action of SSPN overexpression. METHODS The tibialis anterior and quadriceps muscles were isolated from wild-type, mdx, and mdxTG mice and subjected to bulk RNA-Seq and global proteomics analysis using methods to enhance capture of ECM proteins. Data sets were further analyzed through the ingenuity pathway analysis (QIAGEN) and integrative gene set enrichment to identify candidate networks, signaling pathways, and upstream regulators. RESULTS Through our multi-omics approach, we identified 3 classes of differentially expressed genes and proteins in mdxTG muscle, including those that were (1) unrestored (significantly different from wild type, but not from mdx), (2) restored (significantly different from mdx, but not from wild type), and (3) compensatory (significantly different from both wild type and mdx). We identified signaling pathways that may contribute to the rescue phenotype, most notably cytoskeleton and ECM organization pathways. ECM-optimized proteomics revealed an increased abundance of collagens II, V, and XI, along with β-spectrin in mdxTG samples. Using ingenuity pathway analysis, we identified upstream regulators that are computationally predicted to drive compensatory changes, revealing a possible mechanism of SSPN rescue through a rewiring of cell-ECM bidirectional communication. We found that SSPN overexpression results in upregulation of key signaling molecules associated with regulation of cytoskeleton organization and mechanotransduction, including Yap1, Sox9, Rho, RAC, and Wnt. CONCLUSIONS Our findings indicate that SSPN overexpression rescues dystrophin deficiency partially through mechanotransduction signaling cascades mediated through components of the ECM and the cortical cytoskeleton.
Collapse
Affiliation(s)
- Jackie L McCourt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Kristen M Stearns-Reider
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
- Department of Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Hafsa Mamsa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Pranav Kannan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | | | - Cynthia Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Elizabeth M Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Kara M Shin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, CO, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Long S, Xie C, Lu X. Natural polymer‐based adhesive hydrogel for biomedical applications. BIOSURFACE AND BIOTRIBOLOGY 2022. [DOI: 10.1049/bsb2.12036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Siyu Long
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| | - Chaoming Xie
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| | - Xiong Lu
- Key Laboratory of Advanced Technologies of Materials Ministry of Education School of Materials Science and Engineering Southwest Jiaotong University Chengdu China
- Yibin Research Institute Southwest Jiaotong University Yibin China
| |
Collapse
|
14
|
Ghaffari-Bohlouli P, Jafari H, Taebnia N, Abedi A, Amirsadeghi A, Niknezhad SV, Alimoradi H, Jafarzadeh S, Mirzaei M, Nie L, Zhang J, Varma RS, Shavandi A. Protein by-products: Composition, extraction, and biomedical applications. Crit Rev Food Sci Nutr 2022; 63:9436-9481. [PMID: 35546340 DOI: 10.1080/10408398.2022.2067829] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Significant upsurge in animal by-products such as skin, bones, wool, hides, feathers, and fats has become a global challenge and, if not properly disposed of, can spread contamination and viral diseases. Animal by-products are rich in proteins, which can be used as nutritional, pharmacologically functional ingredients, and biomedical materials. Therefore, recycling these abundant and renewable by-products and extracting high value-added components from them is a sustainable approach to reclaim animal by-products while addressing scarce landfill resources. This article appraises the most recent studies conducted in the last five years on animal-derived proteins' separation and biomedical application. The effort encompasses an introduction about the composition, an overview of the extraction and purification methods, and the broad range of biomedical applications of these ensuing proteins.
Collapse
Affiliation(s)
| | - Hafez Jafari
- 3BIO-BioMatter, Faculty of engineering, Free University of Brussels (ULB), Brussels, Belgium
| | - Nayere Taebnia
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ali Abedi
- Department of Life Science Engineering, Faculty of New Sciences and Technology, University of Tehran, Tehran, Iran
| | - Armin Amirsadeghi
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Vahid Niknezhad
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Houman Alimoradi
- School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sina Jafarzadeh
- Department of Energy Conversion and Storage, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mahta Mirzaei
- 3BIO-BioMatter, Faculty of engineering, Free University of Brussels (ULB), Brussels, Belgium
| | - Lei Nie
- 3BIO-BioMatter, Faculty of engineering, Free University of Brussels (ULB), Brussels, Belgium
- College of Life Sciences, Xinyang Normal University, Xinyang, China
| | - Jianye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R. China
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University, Olomouc, Czech Republic
| | - Amin Shavandi
- 3BIO-BioMatter, Faculty of engineering, Free University of Brussels (ULB), Brussels, Belgium
| |
Collapse
|
15
|
Kilmer CE, Walimbe T, Panitch A, Liu JC. Incorporation of a Collagen-Binding Chondroitin Sulfate Molecule to a Collagen Type I and II Blend Hydrogel for Cartilage Tissue Engineering. ACS Biomater Sci Eng 2022; 8:1247-1257. [PMID: 35133126 PMCID: PMC9191256 DOI: 10.1021/acsbiomaterials.1c01248] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adding chondroitin sulfate (CS) to collagen scaffolds has been shown to improve the outcomes for articular cartilage tissue engineering. Instead of physical entrapment or chemical crosslinking of CS within a scaffold, this study investigated the use of CS with attached collagen-binding peptides (termed CS-SILY). This method better recapitulates the aspects of native cartilage while retaining CS within a collagen type I and II blend (Col I/II) hydrogel. CS retention, average fibril diameter, and mechanical properties were altered by varying the number of SILY peptides attached to the CS backbone. When mesenchymal stromal cells (MSCs) were encapsulated within the scaffolds, the addition of CS-SILY molecules resulted in higher sulfated glycosaminoglycan production, and these results suggest that CS-SILY promotes MSC differentiation into chondrocytes. Taken together, our study shows the promise of adding a CS-SILY molecule to a Col I/II hydrogel with encapsulated MSCs to promote cartilage repair.
Collapse
Affiliation(s)
- Claire E Kilmer
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tanaya Walimbe
- School of Biomedical Engineering, University of California Davis, Davis, California 95616, United States
| | - Alyssa Panitch
- School of Biomedical Engineering, University of California Davis, Davis, California 95616, United States.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
16
|
A long-lasting guided bone regeneration membrane from sequentially functionalised photoactive atelocollagen. Acta Biomater 2022; 140:190-205. [PMID: 34896269 DOI: 10.1016/j.actbio.2021.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023]
Abstract
The fast degradation of collagen-based membranes in the biological environment remains a critical challenge, resulting in underperforming Guided Bone Regeneration (GBR) therapy leading to compromised clinical results. Photoactive atelocollagen (AC) systems functionalised with ethylenically unsaturated monomers, such as 4-vinylbenzyl chloride (4VBC), have been shown to generate mechanically competent materials for wound healing, inflammation control and drug delivery, whereby control of the molecular architecture of the AC network is key. Building on this platform, the sequential functionalisation with 4VBC and methacrylic anhydride (MA) was hypothesised to generate UV-cured AC hydrogels with reduced swelling ratio, increased proteolytic stability and barrier functionality for GBR therapy. The sequentially functionalised atelocollagen precursor (SAP) was characterised via TNBS and ninhydrin colourimetric assays, circular dichroism and UV-curing rheometry, which confirmed nearly complete consumption of collagen's primary amino groups, preserved triple helices and fast (< 180 s) gelation kinetics, respectively. Hydrogel's swelling ratio and compression modulus were adjusted depending on the aqueous environment used for UV-curing, whilst the sequential functionalisation of AC successfully generated hydrogels with superior proteolytic stability in vitro compared to both 4VBC-functionalised control and the commercial dental membrane Bio-Gide®. These in vitro results were confirmed in vivo via both subcutaneous implantation and a proof-of-concept study in a GBR calvarial model, indicating integrity of the hydrogel and barrier defect, as well as tissue formation following 1-month implantation in rats. STATEMENT OF SIGNIFICANCE: Collagen-based membranes remain a key component in Guided Bone Regeneration (GBR) therapy, but their properties, e.g. proteolytic stability and soft tissue barrier functionality, are still far from optimal. This is largely attributed to the complex molecular configuration of collagen, which makes chemical accessibility and structure-function relations challenging. Here, we fabricated a UV-cured hydrogel network of atelocollagen, whereby triple helices were sequentially functionalised with two distinct ethylenically unsaturated monomers. The effects of the sequential functionalisation and UV-curing on the macroscopic properties, degradation behaviour and GBR capability were investigated in vitro and in vivo. The results highlight the key role of the sequential functionalisation and provide important insights for the design of future, longer-lasting resorbable membranes for GBR therapy.
Collapse
|
17
|
Hu X, Xia Z, Cai K. Recent advances of 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B 2022; 10:1486-1507. [DOI: 10.1039/d1tb02537f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly recognized as resources for disease treatments and regenerative medicine. Meanwhile, the unique chemical and physical properties of hydrogels provide innate advantages to achieve...
Collapse
|
18
|
The effects of substrate composition and topography on the characteristics and growth of cell cultures of cochlear fibrocytes. Hear Res 2021; 415:108427. [PMID: 34999290 DOI: 10.1016/j.heares.2021.108427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/02/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022]
Abstract
Spiral ligament fibrocytes of the cochlea play homoeostatic roles in hearing and their degeneration contributes to hearing loss. Culturing fibrocytes in vitro provides a way to evaluate their functional characteristics and study possible therapies for hearing loss. We investigated whether in vivo characteristics of fibrocytes could be recapitulated in vitro by modifying the culture substrates and carried out proof of concept studies for potential transplantation of culture cells into the inner ear. Fibrocytes cultured from 4 to 5-week old CD/1 mice were grown on 2D substrates coated with collagen I, II, V or IX and, after harvesting, onto or into 3D substrates (hydrogels) of collagen I alone or mixed collagen I and II at a 1:1 ratio. We also assessed magnetic nanoparticle (MNP) uptake. Cell counts, immunohistochemical and ultrastructural studies showed that fibrocytes grown on 2D substrates proliferated, formed both small spindle-shaped and large flat cells that avidly took up MNPs. Of the different collagen coatings, only collagen II had an effect, causing a reduced size of the larger cells. On hydrogels, the cells were plump/rounded with extended processes, resembling native cells. They formed networks over the surface and became incorporated into the gel. In all culture formats, the majority co-expressed caldesmon, aquaporin 1, S-100 and sodium potassium ATPase, indicating a mixed or uncharacterised phenotype. Time-course experiments showed a decrease to ∼50% of the starting population by 4d after seeding on collagen I hydrogels, but better survival (∼60%) was found on collagen I + II gels, whilst TEM revealed the presence of apoptotic cells. Cells grown within gels additionally showed necrosis. These results demonstrate that fibrocytes grown in 3D recapitulate in vivo morphology of native fibrocytes, but have poorer survival, compared with 2D. Therefore hydrogel cultures could be used to study fibrocyte function and might also offer avenues for cell-replacement therapies, but need more optimization for therapeutic use. Fibrocyte function could be modified using MNPs in combination, for example, with gene transfection.
Collapse
|
19
|
Main and Minor Types of Collagens in the Articular Cartilage: The Role of Collagens in Repair Tissue Evaluation in Chondral Defects. Int J Mol Sci 2021; 22:ijms222413329. [PMID: 34948124 PMCID: PMC8706311 DOI: 10.3390/ijms222413329] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 12/15/2022] Open
Abstract
Several collagen subtypes have been identified in hyaline articular cartilage. The main and most abundant collagens are type II, IX and XI collagens. The minor and less abundant collagens are type III, IV, V, VI, X, XII, XIV, XVI, XXII, and XXVII collagens. All these collagens have been found to play a key role in healthy cartilage, regardless of whether they are more or less abundant. Additionally, an exhaustive evaluation of collagen fibrils in a repaired cartilage tissue after a chondral lesion is necessary to determine the quality of the repaired tissue and even whether or not this repaired tissue is considered hyaline cartilage. Therefore, this review aims to describe in depth all the collagen types found in the normal articular cartilage structure, and based on this, establish the parameters that allow one to consider a repaired cartilage tissue as a hyaline cartilage.
Collapse
|
20
|
Xu Q, Torres JE, Hakim M, Babiak PM, Pal P, Battistoni CM, Nguyen M, Panitch A, Solorio L, Liu JC. Collagen- and hyaluronic acid-based hydrogels and their biomedical applications. MATERIALS SCIENCE & ENGINEERING. R, REPORTS : A REVIEW JOURNAL 2021; 146:100641. [PMID: 34483486 PMCID: PMC8409465 DOI: 10.1016/j.mser.2021.100641] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Hydrogels have been widely investigated in biomedical fields due to their similar physical and biochemical properties to the extracellular matrix (ECM). Collagen and hyaluronic acid (HA) are the main components of the ECM in many tissues. As a result, hydrogels prepared from collagen and HA hold inherent advantages in mimicking the structure and function of the native ECM. Numerous studies have focused on the development of collagen and HA hydrogels and their biomedical applications. In this extensive review, we provide a summary and analysis of the sources, features, and modifications of collagen and HA. Specifically, we highlight the fabrication, properties, and potential biomedical applications as well as promising commercialization of hydrogels based on these two natural polymers.
Collapse
Affiliation(s)
- Qinghua Xu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jessica E Torres
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mazin Hakim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pallabi Pal
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Carly M Battistoni
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Michael Nguyen
- Department of Biomedical Engineering, University of California Davis, Davis, California 95616, United States
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California Davis, Davis, California 95616, United States
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| |
Collapse
|
21
|
Li X, Lu Y, Wang Y, Zhou S, Li L, Zhao F. Thermo-responsive injectable naringin-loaded hydrogel polymerised sodium alginate/bioglass delivery for articular cartilage. Drug Deliv 2021; 28:1290-1300. [PMID: 34176372 PMCID: PMC8238061 DOI: 10.1080/10717544.2021.1938752] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In the human body, joint cartilage is of great importance. It has long been a big therapeutic problem to fix joint cartilage lesions as it appears due to different conditions. Recent stories have shown that the cartilage replacement process must delay the extracellular (ECM) cartilage deterioration and modulate the host's inflammation response. For the reconstruction of the articular cartilage, drug-loaded injectable hydrogels were developed. This hydrogel could retain the chondrocyte phenotype, but the host's inflammatory reaction could also be controlled. The bioglass (BG)/sodium alginate (SA) injectable hydrogels was combined with agarose (AG)/Naringin hydrogel in injectable thermal response for articular cartilage regeneration with a non-chargeable hydrogel that contains both Naringin and BG (Naringin–BG hydrogels). The Naringin–BG hydrogel has an adequate swelling ratio that encourages the fusion of tissue formed with host tissue and enables the gradual release of Naringin bioavailabilities enhanced in situ. The Naringin–BG hydrogel can upgrade the typical chondrocyte phenotype by upregulating aggrecan, SRY-box 9, and collagen type II alpha one chain. It may also stimulate the polarization of M2 macrophage, lower inflammations, and prevent ECM degradations through the decrease of the expressions of the indictable metalloproteinase-13 matrix, nitric oxide synthase, and metalloproteinase-1 matrix. The formed tissues were identical to normal tissues and firmly incorporated with the surrounding tissue after administering the Naringin–BG hydrogels into the rat model articular cartilage defects. Then the injectable Naringin–BG hydrogel increases the bioavailable content of Naringin and retains the chondrocyte phenotype.
Collapse
Affiliation(s)
- Xiang Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yang Lu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yuxin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Shengji Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Liangping Li
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Fengchao Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Wu Z, Korntner SH, Mullen AM, Skoufos I, Tzora A, Zeugolis DI. In the quest of the optimal tissue source (porcine male and female articular, tracheal and auricular cartilage) for the development of collagen sponges for articular cartilage. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
23
|
Han L, Xu N, Lv S, Yin J, Zheng D, Li X. Enhanced In Vitro and In Vivo Efficacy of Alginate/Silk Protein/Hyaluronic Acid with Polypeptide Microsphere Delivery for Tissue Regeneration of Articular Cartilage. J Biomed Nanotechnol 2021; 17:901-909. [PMID: 34082875 DOI: 10.1166/jbn.2021.3071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alginate/Silk fibroin/hyaluronic acid (ALG/SF/HA) nanocomposites were synthesised using blending, inter-linking, and lyophilization methods. We investigated the physicochemical properties of the resulting nanocomposites, including their water retention, weight loss, porosity and cytocompatibility. The optimum ratios of the ALG/SF/HA scaffolding were 3:6.5:0.5. Nanocomposites with optimum ratios were then prepared by integrating pilose antler polypeptides (PAPS) to poly(lactic-co-glycolic acid) (PLGA) microspheres, and the performance was investigated. PAPS-ALG/SF/HA nanocomposites exhibited desirable adhesions and proliferations. Rabbit cartilage deficiencies was developed by the animal model. The cartilage repair effects deficiencies were detected and analyzed between PAPS-SF/ALG/ALG/SF/HA, and control activity classes. The deficiencies were virtually fully remedied after 13 weeks in the presence of PAPS-ALG/SF/HA class, suggesting that the PAPS-ALG/SF/HA nanocomposites had a positive effects on joint cartilage repair.
Collapse
Affiliation(s)
- Long Han
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jianjian Yin
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Dong Zheng
- Department of Orthopedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, China
| | - Xin Li
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| |
Collapse
|
24
|
Xiong X, Liu L, Xu F, Wu X, Yin Z, Dong Y, Qian P. Feprazone Ameliorates TNF-α-Induced Loss of Aggrecan via Inhibition of the SOX-4/ADAMTS-5 Signaling Pathway. ACS OMEGA 2021; 6:7638-7645. [PMID: 33778274 PMCID: PMC7992146 DOI: 10.1021/acsomega.0c06212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/16/2021] [Indexed: 06/12/2023]
Abstract
Background: Arthritis is a cartilage degenerative disease that is mainly induced by the degradation of the cartilage extracellular matrix (ECM), which is found to be regulated by the expression level of a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMT-5), an enzyme degrading Aggrecans in the ECM. Feprazone is a classic nonsteroidal anti-inflammatory drug with promising efficacy in arthritis. The present study aims to investigate the protective effect of Feprazone on the degraded Aggrecan in the human chondrocytes induced with tumor necrosis factor-α (TNF-α) and to clarify the underlying mechanism. Methods: To investigate the effect of Feprazone, the CHON-001 chondrocytes were stimulated with TNF-α (10 ng/mL) in the presence or absence of Feprazone (3, 6 μM) for 24 h. Mitochondrial membrane potential was evaluated using the Rhodamine 123 assay. The gene expressions of interleukin-1β (IL-1β), interleukin-8 (IL-8), monocyte chemotactic protein 1 (MCP-1), and ADAMTS-5 in the treated chondrocytes were detected using real-time quantitative polymerase chain reaction (qRT-PCR), and the protein levels of these targets were determined using enzyme-linked immunosorbent assay (ELISA). SOX-4 was knocked down by transfecting the siRNA into the chondrocytes. Western blot analysis was utilized to evaluate the expression levels of SOX-4, Aggrecan, and protein kinase C (PKCα). Results: First, the reduced mitochondrial membrane potential (ΔΨm) and secretion of proinflammatory factors (IL-1β, IL-8, and MCP-1) induced by TNF-α were significantly reversed by treatment with Feprazone. Second, the expression of Aggrecan was significantly decreased by stimulation with TNF-α via upregulation of ADAMTS-5 but was dramatically reversed by the introduction of Feprazone. Third, we found that TNF-α elevated the expression of ADAMTS-5 by upregulating SOX-4, which was observed to be related to the activation of PKCα. Lastly, the elevated expression of SOX-4 induced by TNF-α was significantly reversed by Feprazone. Conclusions: Feprazone might ameliorate TNF-α-induced loss of Aggrecan via the inhibition of the SOX-4/ADAMTS-5 signaling pathway.
Collapse
|
25
|
Andjelkov N, Riyadh H, Ivarsson M, Kacarevic-Popovic Z, Krstic J, Wretenberg P. The enhancement of cartilage regeneration by use of a chitosan-based scaffold in a 3D model of microfracture in vitro: a pilot evaluation. J Exp Orthop 2021; 8:12. [PMID: 33599885 PMCID: PMC7892646 DOI: 10.1186/s40634-021-00328-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/18/2021] [Indexed: 01/15/2023] Open
Affiliation(s)
- N Andjelkov
- Department of Orthopedics, Västmanlands Regional Hospital, Västerås, Sweden. .,Centre for Clinical Research, Uppsala University, Västmanlands Regional Hospital, Västerås, Sweden. .,Department of Orthopaedics, School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - H Riyadh
- Department of Orthopedics, Västmanlands Regional Hospital, Västerås, Sweden
| | - M Ivarsson
- Department of Health Sciences, University of Örebro, Örebro, Sweden
| | - Z Kacarevic-Popovic
- Department of Radiation Chemistry and Physics, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - J Krstic
- Department of Radiation Chemistry and Physics, Vinca Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - P Wretenberg
- Department of Orthopaedics, School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
26
|
Walker M, Luo J, Pringle EW, Cantini M. ChondroGELesis: Hydrogels to harness the chondrogenic potential of stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111822. [PMID: 33579465 DOI: 10.1016/j.msec.2020.111822] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/01/2023]
Abstract
The extracellular matrix is a highly complex microenvironment, whose various components converge to regulate cell fate. Hydrogels, as water-swollen polymer networks composed by synthetic or natural materials, are ideal candidates to create biologically active substrates that mimic these matrices and target cell behaviour for a desired tissue engineering application. Indeed, the ability to tune their mechanical, structural, and biochemical properties provides a framework to recapitulate native tissues. This review explores how hydrogels have been engineered to harness the chondrogenic response of stem cells for the repair of damaged cartilage tissue. The signalling processes involved in hydrogel-driven chondrogenesis are also discussed, identifying critical pathways that should be taken into account during hydrogel design.
Collapse
Affiliation(s)
- Matthew Walker
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Jiajun Luo
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Eonan William Pringle
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK
| | - Marco Cantini
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, UK.
| |
Collapse
|
27
|
Baumgartner L, Wuertz-Kozak K, Le Maitre CL, Wignall F, Richardson SM, Hoyland J, Ruiz Wills C, González Ballester MA, Neidlin M, Alexopoulos LG, Noailly J. Multiscale Regulation of the Intervertebral Disc: Achievements in Experimental, In Silico, and Regenerative Research. Int J Mol Sci 2021; 22:E703. [PMID: 33445782 PMCID: PMC7828304 DOI: 10.3390/ijms22020703] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a major risk factor of low back pain. It is defined by a progressive loss of the IVD structure and functionality, leading to severe impairments with restricted treatment options due to the highly demanding mechanical exposure of the IVD. Degenerative changes in the IVD usually increase with age but at an accelerated rate in some individuals. To understand the initiation and progression of this disease, it is crucial to identify key top-down and bottom-up regulations' processes, across the cell, tissue, and organ levels, in health and disease. Owing to unremitting investigation of experimental research, the comprehension of detailed cell signaling pathways and their effect on matrix turnover significantly rose. Likewise, in silico research substantially contributed to a holistic understanding of spatiotemporal effects and complex, multifactorial interactions within the IVD. Together with important achievements in the research of biomaterials, manifold promising approaches for regenerative treatment options were presented over the last years. This review provides an integrative analysis of the current knowledge about (1) the multiscale function and regulation of the IVD in health and disease, (2) the possible regenerative strategies, and (3) the in silico models that shall eventually support the development of advanced therapies.
Collapse
Affiliation(s)
- Laura Baumgartner
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY 14623, USA;
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), 81547 Munich, Germany
| | - Christine L. Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK;
| | - Francis Wignall
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Stephen M. Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Judith Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Carlos Ruiz Wills
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Miguel A. González Ballester
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
- Catalan Institution for Research and Advanced Studies (ICREA), Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Jérôme Noailly
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| |
Collapse
|
28
|
Krueger S, Riess A, Jonitz-Heincke A, Weizel A, Seyfarth A, Seitz H, Bader R. Establishment of a New Device for Electrical Stimulation of Non-Degenerative Cartilage Cells In Vitro. Int J Mol Sci 2021; 22:ijms22010394. [PMID: 33401406 PMCID: PMC7794805 DOI: 10.3390/ijms22010394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
In cell-based therapies for cartilage lesions, the main problem is still the formation of fibrous cartilage, caused by underlying de-differentiation processes ex vivo. Biophysical stimulation is a promising approach to optimize cell-based procedures and to adapt them more closely to physiological conditions. The occurrence of mechano-electrical transduction phenomena within cartilage tissue is physiological and based on streaming and diffusion potentials. The application of exogenous electric fields can be used to mimic endogenous fields and, thus, support the differentiation of chondrocytes in vitro. For this purpose, we have developed a new device for electrical stimulation of chondrocytes, which operates on the basis of capacitive coupling of alternating electric fields. The reusable and sterilizable stimulation device allows the simultaneous use of 12 cavities with independently applicable fields using only one main supply. The first parameter settings for the stimulation of human non-degenerative chondrocytes, seeded on collagen type I elastin-based scaffolds, were derived from numerical electric field simulations. Our first results suggest that applied alternating electric fields induce chondrogenic re-differentiation at the gene and especially at the protein level of human de-differentiated chondrocytes in a frequency-dependent manner. In future studies, further parameter optimizations will be performed to improve the differentiation capacity of human cartilage cells.
Collapse
Affiliation(s)
- Simone Krueger
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
- Correspondence: (S.K.); (A.R.)
| | - Alexander Riess
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
- Correspondence: (S.K.); (A.R.)
| | - Anika Jonitz-Heincke
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
| | - Alina Weizel
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
| | - Anika Seyfarth
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
| | - Hermann Seitz
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
| | - Rainer Bader
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
| |
Collapse
|
29
|
Seixas MJ, Martins E, Reis RL, Silva TH. Extraction and Characterization of Collagen from Elasmobranch Byproducts for Potential Biomaterial Use. Mar Drugs 2020; 18:E617. [PMID: 33291538 PMCID: PMC7761862 DOI: 10.3390/md18120617] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
With the worldwide increase of fisheries, fish wastes have had a similar increase, alternatively they can be seen as a source of novel substances for the improvement of society's wellbeing. Elasmobranchs are a subclass fished in high amounts, with some species being mainly bycatch. They possess an endoskeleton composed mainly by cartilage, from which chondroitin sulfate is currently obtained. Their use as a viable source for extraction of type II collagen has been hypothesized with the envisaging of a biomedical application, namely in biomaterials production. In the present work, raw cartilage from shark (Prionace glauca) and ray (Zeachara chilensis and Bathyraja brachyurops) was obtained from a fish processing company and submitted to acidic and enzymatic extractions, to produce acid-soluble collagen (ASC) and pepsin-soluble collagen (PSC). From all the extractions, P. glauca PSC had the highest yield (3.5%), followed by ray ASC (0.92%), ray PSC (0.50%), and P. glauca ASC (0.15%). All the extracts showed similar properties, with the SDS-PAGE profiles being compatible with the presence of both type I and type II collagens. Moreover, the collagen extracts exhibited the competence to maintain their conformation at human basal temperature, presenting a denaturation temperature higher than 37 °C. Hydrogels were produced using P. glauca PSC combined with shark chondroitin sulfate, with the objective of mimicking the human cartilage extracellular matrix. These hydrogels were cohesive and structurally-stable at 37 °C, with rheological measurements exhibiting a conformation of an elastic solid when submitted to shear strain with a frequency up to 4 Hz. This work revealed a sustainable strategy for the valorization of fisheries' by-products, within the concept of a circular economy, consisting of the use of P. glauca, Z. chilensis, and B. brachyurops cartilage for the extraction of collagen, which would be further employed in the development of hydrogels as a proof of concept of its biotechnological potential, ultimately envisaging its use in marine biomaterials to regenerate damaged cartilaginous tissues.
Collapse
Affiliation(s)
- Manuel J. Seixas
- 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; (M.J.S.); or (E.M.); (R.L.R.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Eva Martins
- 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; (M.J.S.); or (E.M.); (R.L.R.)
- ICVS/3B’s–PT Government Associate Laboratory, 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; (M.J.S.); or (E.M.); (R.L.R.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Tiago H. 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; (M.J.S.); or (E.M.); (R.L.R.)
- ICVS/3B’s–PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
30
|
Walimbe T, Panitch A. Best of Both Hydrogel Worlds: Harnessing Bioactivity and Tunability by Incorporating Glycosaminoglycans in Collagen Hydrogels. Bioengineering (Basel) 2020; 7:E156. [PMID: 33276506 PMCID: PMC7711789 DOI: 10.3390/bioengineering7040156] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 01/13/2023] Open
Abstract
Collagen, the most abundant protein in mammals, has garnered the interest of scientists for over 50 years. Its ubiquitous presence in all body tissues combined with its excellent biocompatibility has led scientists to study its potential as a biomaterial for a wide variety of biomedical applications with a high degree of success and widespread clinical approval. More recently, in order to increase their tunability and applicability, collagen hydrogels have frequently been co-polymerized with other natural and synthetic polymers. Of special significance is the use of bioactive glycosaminoglycans-the carbohydrate-rich polymers of the ECM responsible for regulating tissue homeostasis and cell signaling. This review covers the recent advances in the development of collagen-based hydrogels and collagen-glycosaminoglycan blend hydrogels for biomedical research. We discuss the formulations and shortcomings of using collagen in isolation, and the advantages of incorporating glycosaminoglycans (GAGs) in the hydrogels. We further elaborate on modifications used on these biopolymers for tunability and discuss tissue specific applications. The information presented herein will demonstrate the versatility and highly translational value of using collagen blended with GAGs as hydrogels for biomedical engineering applications.
Collapse
Affiliation(s)
- Tanaya Walimbe
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA;
- Department of Surgery, University of California Davis Health, Sacramento, CA 95817, USA
| |
Collapse
|
31
|
Vazquez-Portalatin N, Alfonso-Garcia A, Liu JC, Marcu L, Panitch A. Physical, Biomechanical, and Optical Characterization of Collagen and Elastin Blend Hydrogels. Ann Biomed Eng 2020; 48:2924-2935. [PMID: 32929559 DOI: 10.1007/s10439-020-02605-x] [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: 05/27/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
Collagen and elastin proteins are major components of the extracellular matrix of many organs. The presence of collagen and elastin networks, and their associated properties, in different tissues have led scientists to study collagen and elastin composites for use in tissue engineering. In this study, we characterized physical, biochemical, and optical properties of gels composed of collagen and elastin blends. We demonstrated that the addition of varying amounts of elastin to the constructs alters collagen fibrillogenesis, D-banding pattern length, and storage modulus. However, the addition of elastin does not affect collagen fibril diameter. We also evaluated the autofluorescence properties of the different collagen and elastin blends with fluorescence lifetime imaging (FLIm). Autofluorescence emission showed a red shift with the addition of elastin to the hydrogels. The fluorescence lifetime values of the gels increased with the addition of elastin and were strongly correlated with the storage moduli measurements. These results suggest that FLIm can be used to monitor the gels' mechanical properties nondestructively. These collagen and elastin constructs, along with the FLIm capabilities, can be used to develop and study collagen and elastin composites for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Nelda Vazquez-Portalatin
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA.,Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN, 47907, USA
| | - Alba Alfonso-Garcia
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA
| | - Julie C Liu
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN, 47907, USA.,Davidson School of Chemical Engineering, Purdue University, 480 Stadium Mall Dr, West Lafayette, IN, 47907, USA
| | - Laura Marcu
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA
| | - Alyssa Panitch
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA. .,Department of Surgery, University of California, Davis, 2335 Stockton Boulevard, Sacramento, CA, 95817, USA.
| |
Collapse
|
32
|
Chen L, Liu J, Guan M, Zhou T, Duan X, Xiang Z. Growth Factor and Its Polymer Scaffold-Based Delivery System for Cartilage Tissue Engineering. Int J Nanomedicine 2020; 15:6097-6111. [PMID: 32884266 PMCID: PMC7434569 DOI: 10.2147/ijn.s249829] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/02/2020] [Indexed: 02/05/2023] Open
Abstract
The development of biomaterials, stem cells and bioactive factors has led to cartilage tissue engineering becoming a promising tactic to repair cartilage defects. Various polymer three-dimensional scaffolds that provide an extracellular matrix (ECM) mimicking environment play an important role in promoting cartilage regeneration. In addition, numerous growth factors have been found in the regenerative process. However, it has been elucidated that the uncontrolled delivery of these factors cannot fully exert regenerative potential and can also elicit undesired side effects. Considering the complexity of the ECM, neither scaffolds nor growth factors can independently obtain successful outcomes in cartilage tissue engineering. Therefore, collectively, an appropriate combination of growth factors and scaffolds have great potential to promote cartilage repair effectively; this approach has become an area of considerable interest in recent investigations. Of late, an increasing trend was observed in cartilage tissue engineering towards this combination to develop a controlled delivery system that provides adequate physical support for neo-cartilage formation and also enables spatiotemporally delivery of growth factors to precisely and fully exert their chondrogenic potential. This review will discuss the role of polymer scaffolds and various growth factors involved in cartilage tissue engineering. Several growth factor delivery strategies based on the polymer scaffolds will also be discussed, with examples from recent studies highlighting the importance of spatiotemporal strategies for the controlled delivery of single or multiple growth factors in cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiaxin Liu
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Ming Guan
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Tongqing Zhou
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xin Duan
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Zhou Xiang
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
33
|
Kilmer CE, Battistoni CM, Cox A, Breur GJ, Panitch A, Liu JC. Collagen Type I and II Blend Hydrogel with Autologous Mesenchymal Stem Cells as a Scaffold for Articular Cartilage Defect Repair. ACS Biomater Sci Eng 2020; 6:3464-3476. [PMID: 33463160 PMCID: PMC8287628 DOI: 10.1021/acsbiomaterials.9b01939] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Collagen type II is a promising material to repair cartilage defects since it is a major component of articular cartilage and plays a key role in chondrocyte function. This study investigated the chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (MSCs) embedded within a 3:1 collagen type I to II blend (Col I/II) hydrogel or an all collagen type I (Col I) hydrogel. Glycosaminoglycan (GAG) production in Col I/II hydrogels was statistically higher than that in Col I hydrogels or pellet culture, and these results suggested that adding collagen type II promoted GAG production. Col I/II hydrogels had statistically lower alkaline phosphatase (AP) activity than pellets cultured in a chondrogenic medium. The ability of MSCs encapsulated in Col I/II hydrogels to repair cartilage defects was investigated by creating two cartilage defects in the femurs of rabbits. After 13 weeks, histochemical staining suggested that Col I/II blend hydrogels provided favorable conditions for cartilage repair. Histological scoring revealed a statistically higher cartilage repair score for the Col I/II hydrogels compared to either the Col I hydrogels or empty defect controls. Results from this study suggest that there is clinical value in the cartilage repair capabilities of our Col I/II hydrogel with encapsulated MSCs.
Collapse
Affiliation(s)
- Claire E. Kilmer
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
| | - Carly M. Battistoni
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
| | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University,
West Lafayette, IN, 47907, USA
| | - Gert J. Breur
- Department of Veterinary Clinical Sciences, Purdue
University, West Lafayette, IN, 47907, USA
| | - Alyssa Panitch
- Weldon School of Biomedical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
- School of Biomedical Engineering, University of California
Davis, Davis, CA, 95616, USA
| | - Julie C. Liu
- Davidson School of Chemical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University,
West Lafayette, IN, 47907, USA
| |
Collapse
|
34
|
Abstract
We explore the design and synthesis of hydrogel scaffolds for tissue engineering from the perspective of the underlying polymer chemistry. The key polymers, properties and architectures used, and their effect on tissue growth are discussed.
Collapse
|
35
|
Lee KW, Kuan TC, Lee MW, Yang CS, Hwang LC, Chen CJ, Chang SJ. Effects of bio-mimic collagen II-g-hyaluronic acid copolymers on chondrocyte maintenance. J BIOACT COMPAT POL 2019. [DOI: 10.1177/0883911519876069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Extracellular matrix has an important part of the role in tissue engineering and regenerative medicine, so it is necessary to understand the various interactions between cells and extracellular matrix. Type II collagen and hyaluronic acid are the major structural components of the extracellular matrix of articular cartilage, and they are involved in fibril formation, entanglement and binding. The aim of this study was to prepare type II collagen fibrils with surface grafted with hyaluronic acid modified at the reducing end. The topographic pattern of type II collagen fibrils showed a significant change after the surface coupling of hyaluronic acid according to atomic force microscopy scanning. The presence of hyaluronic acid on the type II collagen fibrillar surface was confirmed by the specific binding of nanogold labelled with lectin. No significant increase in cell proliferation was detected by a WST-1 assay. According to histochemical examination, the maintenance of the round shape of chondrocytes and increased glycosaminoglycan secretion revealed that these cell pellets with Col II- g-hyaluronic acid molecules contained un-dedifferentiated chondrocytes in vitro. In the mixture with the 220-kDa Col II- g-hyaluronic acid copolymer, the expression of type II collagen and aggrecan genes in chondrocytes increased as demonstrated by real-time polymerase chain reaction analysis. Experimental results show that the amount of hyaluronic acid added during culturing of chondrocytes can maintain the functionality of chondrocytes and thus allow for increased cell proliferation that is suitable for tissue repair of human cartilage.
Collapse
Affiliation(s)
- Kuan Wei Lee
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City, Taiwan, ROC
| | - Tang-Ching Kuan
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City, Taiwan, ROC
| | - Ming Wei Lee
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan, ROC
| | - Chen Show Yang
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City, Taiwan, ROC
| | - Lain-Chyr Hwang
- Department of Electrical Engineering, I-Shou University, Kaohsiung City, Taiwan, ROC
| | - Ching-Jung Chen
- School of Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Shwu Jen Chang
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City, Taiwan, ROC
| |
Collapse
|
36
|
Zheng L, Liu S, Cheng X, Qin Z, Lu Z, Zhang K, Zhao J. Intensified Stiffness and Photodynamic Provocation in a Collagen-Based Composite Hydrogel Drive Chondrogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900099. [PMID: 31453055 PMCID: PMC6702628 DOI: 10.1002/advs.201900099] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 06/29/2019] [Indexed: 05/26/2023]
Abstract
Directed differentiation of bone-marrow-derived stem cells (BMSCs) toward chondrogenesis has served as a predominant method for cartilage repair but suffers from poor oriented differentiation tendency and low differentiation efficiency. To overcome these two obstacles, an injectable composite hydrogel that consists of collagen hydrogels serving as the scaffold support to accommodate BMSCs and cadmium selenide (CdSe) quantum dots (QDs) is constructed. The introduction of CdSe QDs considerably strengthens the stiffness of the collagen hydrogels via mutual crosslinking using a natural crosslinker (i.e., genipin), which simultaneously triggers photodynamic provocation (PDP) to produce reactive oxygen species (ROS). Experimental results demonstrate that the intensified stiffness and augmented ROS production can synergistically promote the proliferation of BMSCs, induce cartilage-specific gene expression and increase secretion of glycosaminoglycan. As a result, this approach can facilitate the directed differentiation of BMSCs toward chondrogenesis and accelerate cartilage regeneration in cartilage defect repair, which routes through activation of the TGF-β/SMAD and mTOR signaling pathways, respectively. Thus, this synergistic strategy based on increased stiffness and PDP-mediated ROS production provides a general and instructive approach for developing alternative materials applicable for cartilage repair.
Collapse
Affiliation(s)
- Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
| | - Sijia Liu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
| | - Xiaojing Cheng
- Life Sciences InstituteGuangxi Medical UniversityNo. 22 Shuangyong RoadNanning530021P. R. China
| | - Zainen Qin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
| | - Zhenhui Lu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
| | - Kun Zhang
- Department of Medical UltrasoundShanghai Tenth People's HospitalTongji University School of Medicine301 Yan‐chang‐zhong RoadShanghai200072P. R. China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
- Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
- Department of Orthopaedics Trauma and Hand SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNo. 6 Shuangyong RoadNanning530021P. R. China
| |
Collapse
|
37
|
Walimbe T, Calve S, Panitch A, Sivasankar MP. Incorporation of types I and III collagen in tunable hyaluronan hydrogels for vocal fold tissue engineering. Acta Biomater 2019; 87:97-107. [PMID: 30708064 DOI: 10.1016/j.actbio.2019.01.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 11/28/2022]
Abstract
Vocal fold scarring is the fibrotic manifestation of a variety of voice disorders, and is difficult to treat. Tissue engineering therapies provide a potential strategy to regenerate the native tissue microenvironment in order to restore vocal fold functionality. However, major challenges remain in capturing the complexity of the native tissue and sustaining regeneration. We hypothesized that hydrogels with tunable viscoelastic properties that present relevant biological cues to cells might be better suited as therapeutics. Herein, we characterized the response of human vocal fold fibroblasts to four different biomimetic hydrogels: thiolated hyaluronan (HA) crosslinked with poly(ethylene glycol) diacrylate (PEGDA), HA-PEGDA with type I collagen (HA-Col I), HA-PEGDA with type III collagen (HA-Col III) and HA-PEGDA with type I and III collagen (HA-Col I-Col III). Collagen incorporation allowed for interpenetrating fibrils of collagen within the non-fibrillar HA network, which increased the mechanical properties of the hydrogels. The addition of collagen fibrils also reduced hyaluronidase degradation of HA and hydrogel swelling ratio. Fibroblasts encapsulated in the HA-Col gels adopted a spindle shaped fibroblastic morphology by day 7 and exhibited extensive cytoskeletal networks by day 21, suggesting that the incorporation of collagen was essential for cell adhesion and spreading. Cells remained viable and synthesized new DNA throughout 21 days of culture. Gene expression levels significantly differed between the cells encapsulated in the different hydrogels. Relative fold changes in gene expression of MMP1, COL1A1, fibronectin and decorin suggest higher degrees of remodeling in HA-Col I-Col III gels in comparison to HA-Col I or HA-Col III hydrogels, suggesting that the former may better serve as a natural biomimetic hydrogel for tissue engineering applications. STATEMENT OF SIGNIFICANCE: Voice disorders affect about 1/3rd of the US population and significantly reduce quality of life. Patients with vocal fold fibrosis have few treatment options. Tissue engineering therapies provide a potential strategy to regenerate the native tissue microenvironment in order to restore vocal fold functionality. Various studies have used collagen or thiolated hyaluronan (HA) with gelatin as potential tissue engineering therapies. However, there is room for improvement in providing cells with more relevant biological cues that mimic the native tissue microenvironment and sustain regeneration. The present study introduces the use of type I collagen and type III collagen along with thiolated HA as a natural biomimetic hydrogel for vocal fold tissue engineering applications.
Collapse
Affiliation(s)
- Tanaya Walimbe
- Weldon School of Biomedical Engineering, Purdue University, United States
| | - Sarah Calve
- Weldon School of Biomedical Engineering, Purdue University, United States
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, United States.
| | - M Preeti Sivasankar
- Weldon School of Biomedical Engineering, Purdue University, United States; Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
38
|
Brusatin G, Panciera T, Gandin A, Citron A, Piccolo S. Biomaterials and engineered microenvironments to control YAP/TAZ-dependent cell behaviour. NATURE MATERIALS 2018; 17:1063-1075. [PMID: 30374202 PMCID: PMC6992423 DOI: 10.1038/s41563-018-0180-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/29/2018] [Indexed: 05/11/2023]
Abstract
Mechanical signals are increasingly recognized as overarching regulators of cell behaviour, controlling stemness, organoid biology, tissue development and regeneration. Moreover, aberrant mechanotransduction is a driver of disease, including cancer, fibrosis and cardiovascular defects. A central question remains how cells compute a host of biomechanical signals into meaningful biological behaviours. Biomaterials and microfabrication technologies are essential to address this issue. Here we review a large body of evidence that connects diverse biomaterial-based systems to the functions of YAP/TAZ, two highly related mechanosensitive transcriptional regulators. YAP/TAZ orchestrate the response to a suite of engineered microenviroments, emerging as a universal control system for cells in two and three dimensions, in static or dynamic fashions, over a range of elastic and viscoelastic stimuli, from solid to fluid states. This approach may guide the rational design of technological and material-based platforms with dramatically improved functionalities and inform the generation of new biomaterials for regenerative medicine applications.
Collapse
Affiliation(s)
- Giovanna Brusatin
- Department of Industrial Engineering (DII) and INSTM, University of Padua, Padua, Italy
| | - Tito Panciera
- Department of Molecular Medicine (DMM), University of Padua School of Medicine, Padua, Italy
| | - Alessandro Gandin
- Department of Industrial Engineering (DII) and INSTM, University of Padua, Padua, Italy
- Department of Molecular Medicine (DMM), University of Padua School of Medicine, Padua, Italy
| | - Anna Citron
- Department of Molecular Medicine (DMM), University of Padua School of Medicine, Padua, Italy
| | - Stefano Piccolo
- Department of Molecular Medicine (DMM), University of Padua School of Medicine, Padua, Italy.
- IFOM-the FIRC Institute of Molecular Oncology, .
| |
Collapse
|
39
|
Irawan V, Sung TC, Higuchi A, Ikoma T. Collagen Scaffolds in Cartilage Tissue Engineering and Relevant Approaches for Future Development. Tissue Eng Regen Med 2018; 15:673-697. [PMID: 30603588 PMCID: PMC6250655 DOI: 10.1007/s13770-018-0135-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cartilage tissue engineering (CTE) aims to obtain a structure mimicking native cartilage tissue through the combination of relevant cells, three-dimensional scaffolds, and extraneous signals. Implantation of 'matured' constructs is thus expected to provide solution for treating large injury of articular cartilage. Type I collagen is widely used as scaffolds for CTE products undergoing clinical trial, owing to its ubiquitous biocompatibility and vast clinical approval. However, the long-term performance of pure type I collagen scaffolds would suffer from its limited chondrogenic capacity and inferior mechanical properties. This paper aims to provide insights necessary for advancing type I collagen scaffolds in the CTE applications. METHODS Initially, the interactions of type I/II collagen with CTE-relevant cells [i.e., articular chondrocytes (ACs) and mesenchymal stem cells (MSCs)] are discussed. Next, the physical features and chemical composition of the scaffolds crucial to support chondrogenic activities of AC and MSC are highlighted. Attempts to optimize the collagen scaffolds by blending with natural/synthetic polymers are described. Hybrid strategy in which collagen and structural polymers are combined in non-blending manner is detailed. RESULTS Type I collagen is sufficient to support cellular activities of ACs and MSCs; however it shows limited chondrogenic performance than type II collagen. Nonetheless, type I collagen is the clinically feasible option since type II collagen shows arthritogenic potency. Physical features of scaffolds such as internal structure, pore size, stiffness, etc. are shown to be crucial in influencing the differentiation fate and secreting extracellular matrixes from ACs and MSCs. Collagen can be blended with native or synthetic polymer to improve the mechanical and bioactivities of final composites. However, the versatility of blending strategy is limited due to denaturation of type I collagen at harsh processing condition. Hybrid strategy is successful in maximizing bioactivity of collagen scaffolds and mechanical robustness of structural polymer. CONCLUSION Considering the previous improvements of physical and compositional properties of collagen scaffolds and recent manufacturing developments of structural polymer, it is concluded that hybrid strategy is a promising approach to advance further collagen-based scaffolds in CTE.
Collapse
Affiliation(s)
- Vincent Irawan
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| | - Tzu-Cheng Sung
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Toshiyuki Ikoma
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| |
Collapse
|
40
|
Zhang L, Liu X, Li G, Wang P, Yang Y. Tailoring degradation rates of silk fibroin scaffolds for tissue engineering. J Biomed Mater Res A 2018; 107:104-113. [DOI: 10.1002/jbm.a.36537] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 01/23/2023]
Affiliation(s)
- Luzhong Zhang
- Key Laboratory of Neuroregeneration, Neural Regeneration Co-Innovation Center of Jiangsu Province; Nantong University; Nantong, 226001 People's Republic of China
- Department of Chemistry; Brandeis University; 415 South Street, Waltham Massachusetts, 02454
| | - Xin Liu
- Key Laboratory of Neuroregeneration, Neural Regeneration Co-Innovation Center of Jiangsu Province; Nantong University; Nantong, 226001 People's Republic of China
| | - Guicai Li
- Key Laboratory of Neuroregeneration, Neural Regeneration Co-Innovation Center of Jiangsu Province; Nantong University; Nantong, 226001 People's Republic of China
| | - Peiyuan Wang
- Institute of Imaging, Yantai Affiliated Hospital of Binzhou Medical University; Yantai Shandong People's Republic of China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration, Neural Regeneration Co-Innovation Center of Jiangsu Province; Nantong University; Nantong, 226001 People's Republic of China
| |
Collapse
|
41
|
Injectable and self-crosslinkable hydrogels based on collagen type II and activated chondroitin sulfate for cell delivery. Int J Biol Macromol 2018; 118:2014-2020. [DOI: 10.1016/j.ijbiomac.2018.07.079] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/29/2018] [Accepted: 07/12/2018] [Indexed: 11/24/2022]
|
42
|
杨 波, 常 彦, 凌 鸣, 李 思, 曹 峻. [Demineralized cancellous bone seeded with allogeneic chondrocytes for repairing articular osteochondral defects in rabbits]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1039-1044. [PMID: 30377114 PMCID: PMC6744196 DOI: 10.12122/j.issn.1673-4254.2018.09.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate the effect of demineralized cancellous bone (DCB) seeded with allogeneic chondrocytes for repairing articular osteochondral defects in rabbits. METHODS Articular chondrocytes were isolated from a 1-month-old male New Zealand rabbit for primary culture. The passage 1 chondrocytes were seeded onto prepared rabbit DCB scaffold to construct tissue-engineered cartilage and cultured in vitro for 2 weeks. Full-thickness articular osteochondral defects (3 mm both in diameter and depth) were created on both sides of the femoral medial condyles in 30 New Zealand white rabbits (age 4- 5 months). In 20 of the rabbits, the defects were filled with the tissue-engineered cartilage on the right side (group A) and with DCB only on the left side (group B); the remaining 10 rabbits did not receive any implantation in the defects to serve as the control (group C). At 1, 3, and 6 months after the implantation, tissue samples were collected from the defects for macroscopic observation and histological examination with Toluidine blue (TB) and collagen type Ⅱ staining. The effect of defect repair using the tissue-engineered cartilage was assessed at 6 months based on the histological scores. RESULTS The prepared DCB had a spongy 3D structure with open and interconnected micropores of various sizes and showed good plasticity and mechanical strength. DCB began to degrade within 1 month after implantation and was totally absorbed at 3 months. At 6 months after implantation, the defects filled with the chondrocyte-seeded DCB were repaired mainly by hyaline-like cartilage tissues, which were well integrated to the adjacent cartilage without clear boundaries and difficult to recognize. The chondrocytes were located in the lacunate and arranged in vertical columns in the deep repaired tissue, where matrix proteoglycans and collagen type Ⅱ were distributed homogeneously close to the normal cartilage. The subchondral bone plate was reconstructed completely. The defects implanted with DCB only were filled with fibrocartilage tissue, as compared with fibrous tissue in the control defects. The histological scores in group A were significantly superior to those in group B and C (P < 0.05), but the scores for subchondral bone plate reconstruction were comparable between groups A and B at 6 months. CONCLUSIONS DCB is a good scaffold material for preparing tissue-engineered cartilage, and chondrocyte- seeded DCB can repair articular osteochondral defects by inducing the generation of hayline-like cartilage.
Collapse
Affiliation(s)
- 波 杨
- 陕西省人民医院骨科//西安交通大学第三附属医院,陕西 西安 710068Department of Orthopedics, Shannxi Provincial People's Hospital/Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, China
| | - 彦海 常
- 陕西省人民医院骨科//西安交通大学第三附属医院,陕西 西安 710068Department of Orthopedics, Shannxi Provincial People's Hospital/Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, China
| | - 鸣 凌
- 陕西省人民医院骨科//西安交通大学第三附属医院,陕西 西安 710068Department of Orthopedics, Shannxi Provincial People's Hospital/Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710068, China
| | - 思远 李
- 西安交通大学第二附属医院麻醉科,陕西 西安 710004Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - 峻岭 曹
- 西安交通大学医学部地方病研究所,环境与疾病相关基因教育部重点实验室,陕西 西安 710061Xi'an Jiaotong University Health Science Center, Institute of Endemic Diseases, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, China
| |
Collapse
|
43
|
Wang KH, Wan R, Chiu LH, Tsai YH, Fang CL, Bowley JF, Chen KC, Shih HN, Lai WFT. Effects of collagen matrix and bioreactor cultivation on cartilage regeneration of a full-thickness critical-size knee joint cartilage defects with subchondral bone damage in a rabbit model. PLoS One 2018; 13:e0196779. [PMID: 29746554 PMCID: PMC5945026 DOI: 10.1371/journal.pone.0196779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 04/19/2018] [Indexed: 12/23/2022] Open
Abstract
Cartilage has limited self-repair ability. The purpose of this study was to investigate the effects of different species of collagen-engineered neocartilage for the treatment of critical-size defects in the articular joint in a rabbit model. Type II and I collagen obtained from rabbits and rats was mixed to form a scaffold. The type II/I collagen scaffold was then mixed with rabbit chondrocytes to biofabricate neocartilage constructs using a rotating cell culture system [three-dimensional (3D)-bioreactor]. The rabbit chondrocytes were mixed with rabbit collagen scaffold and rat collagen scaffold to form neoRBT (neo-rabbit cartilage) and neoRAT (neo-rat cartilage) constructs, respectively. The neocartilage matrix constructs were implanted into surgically created defects in rabbit knee chondyles, and histological examinations were performed after 2 and 3 months. Cartilage-like lacunae formation surrounding the chondrocytes was noted in the cell cultures. After 3 months, both the neoRBT and neoRAT groups showed cartilage-like repair tissue covering the 5-mm circular, 4-mm-deep defects that were created in the rabbit condyle and filled with neocartilage plugs. Reparative chondrocytes were aligned as apparent clusters in both the neoRAT and neoRBT groups. Both neoRBT and neoRAT cartilage repair demonstrated integration with healthy adjacent tissue; however, more integration was obtained using the neoRAT cartilage. Our data indicate that different species of type II/I collagen matrix and 3D bioreactor cultivation can facilitate cartilage engineering in vitro for the repair of critical-size defect.
Collapse
Affiliation(s)
- Kuo-Hwa Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
- Department of Obstetrics and Gynecology, Chung Kang branch, Cheng Ching Hospital, Taichung, Taiwan, ROC
| | - Richard Wan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
| | - Li-Hsuan Chiu
- Department of Research, Taipei Medical University-Shaung-Ho Hospital, Taipei, Taiwan, ROC
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States of America
| | - Yu-Hui Tsai
- Department of Research, Taipei Medical University-Shaung-Ho Hospital, Taipei, Taiwan, ROC
| | - Chia-Lang Fang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - John F. Bowley
- Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Kuan-Chou Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
| | - Hsin-Nung Shih
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Chang Gung University, Linkou Taoyuan, Taiwan, ROC
- * E-mail: (HNS); (WFTL)
| | - Wen-Fu Thomas Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, ROC
- Department of Research, Taipei Medical University-Shaung-Ho Hospital, Taipei, Taiwan, ROC
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, United States of America
- * E-mail: (HNS); (WFTL)
| |
Collapse
|
44
|
Yu F, Zong C, Jin S, Zheng J, Chen N, Huang J, Chen Y, Huang F, Yang Z, Tang Y, Ding G. Optimization of Extraction Conditions and Characterization of Pepsin-Solubilised Collagen from Skin of Giant Croaker (Nibea japonica). Mar Drugs 2018; 16:md16010029. [PMID: 29342895 PMCID: PMC5793077 DOI: 10.3390/md16010029] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/06/2018] [Accepted: 01/10/2018] [Indexed: 12/22/2022] Open
Abstract
In the present study, response surface methodology was performed to investigate the effects of extraction parameters on pepsin-solubilised collagen (PSC) from the skin of the giant croaker Nibea japonica. The optimum extraction conditions of PSC were as follows: concentration of pepsin was 1389 U/g, solid-liquid ratio was 1:57 and hydrolysis time was 8.67 h. Under these conditions, the extraction yield of PSC was up to 84.85%, which is well agreement with the predict value of 85.03%. The PSC from Nibea japonica skin was then characterized as type I collagen by using sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). The fourier transforms infrared spetroscopy (FTIR) analysis revealed that PSC maintains its triple-helical structure by the hydrogen bond. All PSCs were soluble in the pH range of 1.0-4.0 and decreases in solubility were observed at neutral or alkaline conditions. All PSCs had a decrease in solubility in the presence of sodium chloride, especially with a concentration above 2%. So, the Nibea japonica skin could serve as another potential source of collagen.
Collapse
Affiliation(s)
- Fangmiao Yu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Chuhong Zong
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Shujie Jin
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Jiawen Zheng
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Nan Chen
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Ju Huang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Yan Chen
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Fangfang Huang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Zuisu Yang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Yunping Tang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| | - Guofang Ding
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China.
| |
Collapse
|
45
|
Sánchez-Téllez DA, Téllez-Jurado L, Rodríguez-Lorenzo LM. Hydrogels for Cartilage Regeneration, from Polysaccharides to Hybrids. Polymers (Basel) 2017; 9:E671. [PMID: 30965974 PMCID: PMC6418920 DOI: 10.3390/polym9120671] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022] Open
Abstract
The aims of this paper are: (1) to review the current state of the art in the field of cartilage substitution and regeneration; (2) to examine the patented biomaterials being used in preclinical and clinical stages; (3) to explore the potential of polymeric hydrogels for these applications and the reasons that hinder their clinical success. The studies about hydrogels used as potential biomaterials selected for this review are divided into the two major trends in tissue engineering: (1) the use of cell-free biomaterials; and (2) the use of cell seeded biomaterials. Preparation techniques and resulting hydrogel properties are also reviewed. More recent proposals, based on the combination of different polymers and the hybridization process to improve the properties of these materials, are also reviewed. The combination of elements such as scaffolds (cellular solids), matrices (hydrogel-based), growth factors and mechanical stimuli is needed to optimize properties of the required materials in order to facilitate tissue formation, cartilage regeneration and final clinical application. Polymer combinations and hybrids are the most promising materials for this application. Hybrid scaffolds may maximize cell growth and local tissue integration by forming cartilage-like tissue with biomimetic features.
Collapse
Affiliation(s)
- Daniela Anahí Sánchez-Téllez
- Instituto Politécnico Nacional-ESIQIE, Depto. Ing. en Metalurgia y Materiales, UPALM-Zacatenco, Mexico City 07738, Mexico.
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain.
| | - Lucía Téllez-Jurado
- Instituto Politécnico Nacional-ESIQIE, Depto. Ing. en Metalurgia y Materiales, UPALM-Zacatenco, Mexico City 07738, Mexico.
| | - Luís María Rodríguez-Lorenzo
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain.
- Department Polymeric Nanomaterials and Biomaterials, ICTP-CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| |
Collapse
|
46
|
Ayerst BI, Merry CLR, Day AJ. The Good the Bad and the Ugly of Glycosaminoglycans in Tissue Engineering Applications. Pharmaceuticals (Basel) 2017; 10:E54. [PMID: 28608822 PMCID: PMC5490411 DOI: 10.3390/ph10020054] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022] Open
Abstract
High sulfation, low cost, and the status of heparin as an already FDA- and EMA- approved product, mean that its inclusion in tissue engineering (TE) strategies is becoming increasingly popular. However, the use of heparin may represent a naïve approach. This is because tissue formation is a highly orchestrated process, involving the temporal expression of numerous growth factors and complex signaling networks. While heparin may enhance the retention and activity of certain growth factors under particular conditions, its binding 'promiscuity' means that it may also inhibit other factors that, for example, play an important role in tissue maintenance and repair. Within this review we focus on articular cartilage, highlighting the complexities and highly regulated processes that are involved in its formation, and the challenges that exist in trying to effectively engineer this tissue. Here we discuss the opportunities that glycosaminoglycans (GAGs) may provide in advancing this important area of regenerative medicine, placing emphasis on the need to move away from the common use of heparin, and instead focus research towards the utility of specific GAG preparations that are able to modulate the activity of growth factors in a more controlled and defined manner, with less off-target effects.
Collapse
Affiliation(s)
- Bethanie I Ayerst
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| | - Catherine L R Merry
- Stem Cell Glycobiology Group, Wolfson Centre for Stem Cells, Tissue Engineering & Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| |
Collapse
|