1
|
Giang HN, Le ATK, Huynh TNA, Phung TK, Sakai W. Effect of additives on fabrication and properties of hydroxypropyl methylcellulose-based hydrogels. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04610-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
2
|
Demarquay C, Moussa L, Réthoré G, Milliat F, Weiss P, Mathieu N. Embedding MSCs in Si-HPMC hydrogel decreased MSC-directed host immune response and increased the regenerative potential of macrophages. Regen Biomater 2022; 9:rbac022. [PMID: 35784096 PMCID: PMC9245650 DOI: 10.1093/rb/rbac022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/22/2022] [Accepted: 04/10/2022] [Indexed: 11/14/2022] Open
Abstract
Embedding mesenchymal stromal cells (MSCs) in biomaterial is a subject of increasing interest in the field of Regenerative Medicine. Speeding up the clinical use of MSCs is dependent on the use of non-syngeneic models in accordance with Good Manufacturing Practices (GMP) requirements and on costs. To this end, in this study, we analyzed the in vivo host immune response following local injection of silanized hydroxypropyl methylcellulose (Si-HPMC)-embedded human MSCs in a rat model developing colorectal damage induced by ionizing radiation. Plasma and lymphocytes from mesenteric lymph nodes were harvested in addition to colonic tissue. We set up tests, using flow cytometry and a live imaging system, to highlight the response to specific antibodies and measure the cytotoxicity of lymphocytes against injected MSCs. We demonstrated that Si-HPMC protects MSCs from specific antibodies production and from apoptosis by lymphocytes. We also observed that Si-HPMC does not modify innate immune response infiltrate in vivo, and that in vitro co-culture of Si-HPMC-embedded MSCs impacts macrophage inflammatory response depending on the microenvironment but, more importantly, increases the macrophage regenerative response through Wnt-family and VEGF gene expression. This study furthers our understanding of the mechanisms involved, with a view to improving the therapeutic benefits of biomaterial-assisted cell therapy by modulating the host immune response. The decrease in specific immune response against injected MSCs protected by Si-HPMC also opens up new possibilities for allogeneic clinical use.
Collapse
Affiliation(s)
- Christelle Demarquay
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, Fontenay-aux-Roses 92262, France
| | - Lara Moussa
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, Fontenay-aux-Roses 92262, France
| | - Gildas Réthoré
- Faculté de Chirurgie Dentaire, Regenerative Medicine and Skeleton (RMeS) Laboratory, Université de Nantes, Nantes 44042, France
| | - Fabien Milliat
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, Fontenay-aux-Roses 92262, France
| | - Pierre Weiss
- Faculté de Chirurgie Dentaire, Regenerative Medicine and Skeleton (RMeS) Laboratory, Université de Nantes, Nantes 44042, France
| | | |
Collapse
|
3
|
Szustak M, Gendaszewska-Darmach E. Nanocellulose-Based Scaffolds for Chondrogenic Differentiation and Expansion. Front Bioeng Biotechnol 2021; 9:736213. [PMID: 34485266 PMCID: PMC8415884 DOI: 10.3389/fbioe.2021.736213] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Nanocellulose deserves special attention among the large group of biocompatible biomaterials. It exhibits good mechanical properties, which qualifies it for potential use as a scaffold imitating cartilage. However, the reconstruction of cartilage is a big challenge due to this tissue's limited regenerative capacity resulting from its lack of vascularization, innervations, and sparsely distributed chondrocytes. This feature restricts the infiltration of progenitor cells into damaged sites. Unfortunately, differentiated chondrocytes are challenging to obtain, and mesenchymal stem cells have become an alternative approach to promote chondrogenesis. Importantly, nanocellulose scaffolds induce the differentiation of stem cells into chondrocyte phenotypes. In this review, we present the recent progress of nanocellulose-based scaffolds promoting the development of cartilage tissue, especially within the emphasis on chondrogenic differentiation and expansion.
Collapse
Affiliation(s)
| | - Edyta Gendaszewska-Darmach
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Lodz, Poland
| |
Collapse
|
4
|
Binder H, Hoffman L, Zak L, Tiefenboeck T, Aldrian S, Albrecht C. Clinical evaluation after matrix-associated autologous chondrocyte transplantation : a comparison of four different graft types. Bone Joint Res 2021; 10:370-379. [PMID: 34189928 PMCID: PMC8333036 DOI: 10.1302/2046-3758.107.bjr-2020-0370.r1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aims The aim of this retrospective study was to determine if there are differences in short-term clinical outcomes among four different types of matrix-associated autologous chondrocyte transplantation (MACT). Methods A total of 88 patients (mean age 34 years (SD 10.03), mean BMI 25 kg/m2 (SD 3.51)) with full-thickness chondral lesions of the tibiofemoral joint who underwent MACT were included in this study. Clinical examinations were performed preoperatively and 24 months after transplantation. Clinical outcomes were evaluated using the International Knee Documentation Committee (IKDC) Subjective Knee Form, the Brittberg score, the Tegner Activity Scale, and the visual analogue scale (VAS) for pain. The Kruskal-Wallis test by ranks was used to compare the clinical scores of the different transplant types. Results The mean defect size of the tibiofemoral joint compartment was 4.28 cm2 (SD 1.70). In total, 11 patients (12.6%) underwent transplantation with Chondro-Gide (matrix-associated autologous chondrocyte implantation (MACI)), 40 patients (46.0%) with Hyalograft C (HYAFF), 21 patients (24.1%) with Cartilage Regeneration System (CaReS), and 15 patients (17.2%) with NOVOCART 3D. The mean IKDC Subjective Knee Form score improved from 35.71 (SD 6.44) preoperatively to 75.26 (SD 18.36) after 24 months postoperatively in the Hyalograft group, from 35.94 (SD 10.29) to 71.57 (SD 16.31) in the Chondro-Gide (MACI) group, from 37.06 (SD 5.42) to 71.49 (SD 6.76) in the NOVOCART 3D group, and from 45.05 (SD 15.83) to 70.33 (SD 19.65) in the CaReS group. Similar improvements were observed in the VAS and Brittberg scores. Conclusion Two years postoperatively, there were no significant differences in terms of outcomes. Our data demonstrated that MACT, regardless of the implants used, resulted in good clinical improvement two years after transplantation for localized tibiofemoral defects. Cite this article: Bone Joint Res 2021;10(7):370–379.
Collapse
Affiliation(s)
- Harald Binder
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Lukas Hoffman
- First Orthopedic Department, Orthopedic Hospital Vienna Speising, Vienna, Austria
| | - Lukas Zak
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Tiefenboeck
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Silke Aldrian
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Christian Albrecht
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria.,First Orthopedic Department, Orthopedic Hospital Vienna Speising, Vienna, Austria
| |
Collapse
|
5
|
Gao Y, Peng K, Mitragotri S. Covalently Crosslinked Hydrogels via Step-Growth Reactions: Crosslinking Chemistries, Polymers, and Clinical Impact. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006362. [PMID: 33988273 DOI: 10.1002/adma.202006362] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/24/2020] [Indexed: 06/12/2023]
Abstract
Hydrogels are an important class of biomaterials with the unique property of high-water content in a crosslinked polymer network. In particular, chemically crosslinked hydrogels have made a great clinical impact in past years because of their desirable mechanical properties and tunability of structural and chemical properties. Various polymers and step-growth crosslinking chemistries are harnessed for fabricating such covalently crosslinked hydrogels for translational research. However, selecting appropriate crosslinking chemistries and polymers for the intended clinical application is time-consuming and challenging. It requires the integration of polymer chemistry knowledge with thoughtful crosslinking reaction design. This task becomes even more challenging when other factors such as the biological mechanisms of the pathology, practical administration routes, and regulatory requirements add additional constraints. In this review, key features of crosslinking chemistries and polymers commonly used for preparing translatable hydrogels are outlined and their performance in biological systems is summarized. The examples of effective polymer/crosslinking chemistry combinations that have yielded clinically approved hydrogel products are specifically highlighted. These hydrogel design parameters in the context of the regulatory process and clinical translation barriers, providing a guideline for the rational selection of polymer/crosslinking chemistry combinations to construct hydrogels with high translational potential are further considered.
Collapse
Affiliation(s)
- Yongsheng Gao
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering, Boston, MA, 02115, USA
| | - Kevin Peng
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering, Boston, MA, 02115, USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute of Biologically Inspired Engineering, Boston, MA, 02115, USA
| |
Collapse
|
6
|
Tourné-Péteilh C, Barège M, Lions M, Martinez J, Devoisselle JM, Aubert-Pouessel A, Subra G, Mehdi A. Encapsulation of BSA in hybrid PEG hydrogels: stability and controlled release. RSC Adv 2021; 11:30887-30897. [PMID: 35498928 PMCID: PMC9041318 DOI: 10.1039/d1ra03547a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/05/2021] [Indexed: 11/23/2022] Open
Abstract
Hybrid hydrogels based on silylated polyethylene glycol, Si-PEG, were evaluated as hybrid matrices able to trap, stabilize and release bovine serum albumin (BSA) in a controlled manner. Parameters of the inorganic condensation reaction leading to a siloxane (Si–O–Si) three dimensional network were carefully investigated, in particular the temperature, the surrounding hygrometry and the Si-PEG concentration. The resulting hydrogel structural features affected the stability, swelling, and mechanical properties of the network, leading to different protein release profiles. Elongated polymer assemblies were observed, the length of which ranged from 150 nm to over 5 μm. The length could be correlated to the Si–O–Si condensation rate from 60% (hydrogels obtained at 24 °C) to about 90% (xerogels obtained at 24 °C), respectively. Consequently, the controlled release of BSA could be achieved from hours to several weeks, with respect to the fibers' length and the condensation rate. The protein stability was evaluated by means of a thermal study. The main results gave insight into the biomolecule structure preservation during polymerisation, with ΔG < 0 for encapsulated BSA in any conditions, below the melting temperature (65 °C). Silylated hybrid hydrogels of polyethylene glycol were designed to trap, stabilize and release a model protein (bovine serum albumin). Fine-tuning sol–gel reactions lead to sustained release of BSA over weeks, with good insight of protein stability.![]()
Collapse
Affiliation(s)
| | - Maeva Barège
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Mathieu Lions
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean Martinez
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | - Gilles Subra
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Ahmad Mehdi
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| |
Collapse
|
7
|
Abbass MMS, El-Rashidy AA, Sadek KM, Moshy SE, Radwan IA, Rady D, Dörfer CE, Fawzy El-Sayed KM. Hydrogels and Dentin-Pulp Complex Regeneration: From the Benchtop to Clinical Translation. Polymers (Basel) 2020; 12:E2935. [PMID: 33316886 PMCID: PMC7763835 DOI: 10.3390/polym12122935] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Dentin-pulp complex is a term which refers to the dental pulp (DP) surrounded by dentin along its peripheries. Dentin and dental pulp are highly specialized tissues, which can be affected by various insults, primarily by dental caries. Regeneration of the dentin-pulp complex is of paramount importance to regain tooth vitality. The regenerative endodontic procedure (REP) is a relatively current approach, which aims to regenerate the dentin-pulp complex through stimulating the differentiation of resident or transplanted stem/progenitor cells. Hydrogel-based scaffolds are a unique category of three dimensional polymeric networks with high water content. They are hydrophilic, biocompatible, with tunable degradation patterns and mechanical properties, in addition to the ability to be loaded with various bioactive molecules. Furthermore, hydrogels have a considerable degree of flexibility and elasticity, mimicking the cell extracellular matrix (ECM), particularly that of the DP. The current review presents how for dentin-pulp complex regeneration, the application of injectable hydrogels combined with stem/progenitor cells could represent a promising approach. According to the source of the polymeric chain forming the hydrogel, they can be classified into natural, synthetic or hybrid hydrogels, combining natural and synthetic ones. Natural polymers are bioactive, highly biocompatible, and biodegradable by naturally occurring enzymes or via hydrolysis. On the other hand, synthetic polymers offer tunable mechanical properties, thermostability and durability as compared to natural hydrogels. Hybrid hydrogels combine the benefits of synthetic and natural polymers. Hydrogels can be biofunctionalized with cell-binding sequences as arginine-glycine-aspartic acid (RGD), can be used for local delivery of bioactive molecules and cellularized with stem cells for dentin-pulp regeneration. Formulating a hydrogel scaffold material fulfilling the required criteria in regenerative endodontics is still an area of active research, which shows promising potential for replacing conventional endodontic treatments in the near future.
Collapse
Affiliation(s)
- Marwa M. S. Abbass
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (M.M.S.A.); (S.E.M.); (I.A.R.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
| | - Aiah A. El-Rashidy
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
- Biomaterials Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Khadiga M. Sadek
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
- Biomaterials Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Sara El Moshy
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (M.M.S.A.); (S.E.M.); (I.A.R.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
| | - Israa Ahmed Radwan
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (M.M.S.A.); (S.E.M.); (I.A.R.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
| | - Dina Rady
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (M.M.S.A.); (S.E.M.); (I.A.R.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (A.A.E.-R.); (K.M.S.)
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
8
|
Réthoré G, Boyer C, Kouadio K, Toure A, Lesoeur J, Halgand B, Jordana F, Guicheux J, Weiss P. Silanization of Chitosan and Hydrogel Preparation for Skeletal Tissue Engineering. Polymers (Basel) 2020; 12:polym12122823. [PMID: 33261192 PMCID: PMC7761294 DOI: 10.3390/polym12122823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering is a multidisciplinary field that relies on the development of customized biomaterial to support cell growth, differentiation and matrix production. Toward that goal, we designed the grafting of silane groups onto the chitosan backbone (Si-chito) for the preparation of in situ setting hydrogels in association with silanized hydroxypropyl methylcellulose (Si-HPMC). Once functionalized, the chitosan was characterized, and the presence of silane groups and its ability to gel were demonstrated by rheology that strongly suggests the presence of silane groups. Throughout physicochemical investigations, the Si-HPMC hydrogels containing Si-chito were found to be stiffer with an injection force unmodified. The presence of chitosan within the hydrogel has demonstrated a higher adhesion of the hydrogel onto the surface of tissues. The results of cell viability assays indicated that there was no cytotoxicity of Si-chito hydrogels in 2D and 3D culture of human SW1353 cells and human adipose stromal cells, respectively. Moreover, Si-chito allows the transplantation of human nasal chondrocytes in the subcutis of nude mice while maintaining their viability and extracellular matrix secretory activity. To conclude, Si-chito mixed with Si-HPMC is an injectable, self-setting and cytocompatible hydrogel able to support the in vitro and in vivo viability and activity of hASC.
Collapse
Affiliation(s)
- Gildas Réthoré
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
- CHU Nantes, PHU4 OTONN, F-44093 Nantes, France
| | - Cécile Boyer
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
| | - Kouakou Kouadio
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
| | - Amadou Toure
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
- Department of Odontology, Faculty of Medicine, Pharmacy and Odontology, University Cheikh Anta DIOP, 12500 Dakar, Senegal
| | - Julie Lesoeur
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
| | - Boris Halgand
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
- CHU Nantes, PHU4 OTONN, F-44093 Nantes, France
| | - Fabienne Jordana
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
- CHU Nantes, PHU4 OTONN, F-44093 Nantes, France
| | - Jérôme Guicheux
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
- CHU Nantes, PHU4 OTONN, F-44093 Nantes, France
| | - Pierre Weiss
- Dental Faculty, Université de Nantes, UMR 1229, RMeS, Regenerative Medicine and Skeleton, INSERM, ONIRIS, F-44042 Nantes, France; (G.R.); (C.B.); (K.K.); (A.T.); (J.L.); (B.H.); (F.J.); (J.G.)
- Institut National de la Santé et de la Recherche Médicale, Université de Nantes, UFR Odontologie, F-44042 Nantes, France
- CHU Nantes, PHU4 OTONN, F-44093 Nantes, France
- Correspondence:
| |
Collapse
|
9
|
Flegeau K, Toquet C, Rethore G, d'Arros C, Messager L, Halgand B, Dupont D, Autrusseau F, Lesoeur J, Veziers J, Bordat P, Bresin A, Guicheux J, Delplace V, Gautier H, Weiss P. In Situ Forming, Silanized Hyaluronic Acid Hydrogels with Fine Control Over Mechanical Properties and In Vivo Degradation for Tissue Engineering Applications. Adv Healthc Mater 2020; 9:e2000981. [PMID: 32864869 DOI: 10.1002/adhm.202000981] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/09/2020] [Indexed: 12/19/2022]
Abstract
In situ forming hydrogels that can be injected into tissues in a minimally-invasive fashion are appealing as delivery vehicles for tissue engineering applications. Ideally, these hydrogels should have mechanical properties matching those of the host tissue, and a rate of degradation adapted for neo-tissue formation. Here, the development of in situ forming hyaluronic acid hydrogels based on the pH-triggered condensation of silicon alkoxide precursors into siloxanes is reported. Upon solubilization and pH adjustment, the low-viscosity precursor solutions are easily injectable through fine-gauge needles prior to in situ gelation. Tunable mechanical properties (stiffness from 1 to 40 kPa) and associated tunable degradability (from 4 days to more than 3 weeks in vivo) are obtained by varying the degree of silanization (from 4.3% to 57.7%) and molecular weight (120 and 267 kDa) of the hyaluronic acid component. Following cell encapsulation, high cell viability (> 80%) is obtained for at least 7 days. Finally, the in vivo biocompatibility of silanized hyaluronic acid gels is verified in a subcutaneous mouse model and a relationship between the inflammatory response and the crosslink density is observed. Silanized hyaluronic acid hydrogels constitute a tunable hydrogel platform for material-assisted cell therapies and tissue engineering applications.
Collapse
Affiliation(s)
- Killian Flegeau
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Claire Toquet
- Department of Pathology University Hospital of Nantes Nantes F‐44042 France
| | - Gildas Rethore
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Cyril d'Arros
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Léa Messager
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Boris Halgand
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Davy Dupont
- HTL S.A.S 7 Rue Alfred Kastler Javené 35133 France
| | - Florent Autrusseau
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Julie Lesoeur
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- SC3M SFR Santé F. Bonamy FED 4203 UMS Inserm 016 CNRS 3556 Nantes F‐44042 France
| | - Joëlle Veziers
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
- SC3M SFR Santé F. Bonamy FED 4203 UMS Inserm 016 CNRS 3556 Nantes F‐44042 France
| | | | | | - Jérôme Guicheux
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| | - Vianney Delplace
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
| | - Hélène Gautier
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- Université de Nantes Faculté de Pharmacie Laboratoire de Pharmacie Galénique Nantes F‐44042 France
| | - Pierre Weiss
- Université de Nantes ONIRIS INSERM Regenerative Medicine and Skeleton, RMeS, UMR 1229 1 Pl A Ricordeau Nantes F‐44042 France
- UFR Odontologie Université de Nantes Nantes F‐44042 France
- CHU Nantes PHU4 OTONN Nantes F‐44042 France
| |
Collapse
|
10
|
Li J, Wu C, Chu PK, Gelinsky M. 3D printing of hydrogels: Rational design strategies and emerging biomedical applications. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2020; 140:100543. [DOI: 10.1016/j.mser.2020.100543] [Citation(s) in RCA: 350] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Boyer C, Réthoré G, Weiss P, d’Arros C, Lesoeur J, Vinatier C, Halgand B, Geffroy O, Fusellier M, Vaillant G, Roy P, Gauthier O, Guicheux J. A Self-Setting Hydrogel of Silylated Chitosan and Cellulose for the Repair of Osteochondral Defects: From in vitro Characterization to Preclinical Evaluation in Dogs. Front Bioeng Biotechnol 2020; 8:23. [PMID: 32117912 PMCID: PMC7025592 DOI: 10.3389/fbioe.2020.00023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage (AC) may be affected by many injuries including traumatic lesions that predispose to osteoarthritis. Currently there is no efficient cure for cartilage lesions. In that respect, new strategies for regenerating AC are contemplated with interest. In this context, we aim to develop and characterize an injectable, self-hardening, mechanically reinforced hydrogel (Si-HPCH) composed of silanised hydroxypropymethyl cellulose (Si-HPMC) mixed with silanised chitosan. The in vitro cytocompatibility of Si-HPCH was tested using human adipose stromal cells (hASC). In vivo, we first mixed Si-HPCH with hASC to observe cell viability after implantation in nude mice subcutis. Si-HPCH associated or not with canine ASC (cASC), was then tested for the repair of osteochondral defects in canine femoral condyles. Our data demonstrated that Si-HPCH supports hASC viability in culture. Moreover, Si-HPCH allows the transplantation of hASC in the subcutis of nude mice while maintaining their viability and secretory activity. In the canine osteochondral defect model, while the empty defects were only partially filled with a fibrous tissue, defects filled with Si-HPCH with or without cASC, revealed a significant osteochondral regeneration. To conclude, Si-HPCH is an injectable, self-setting and cytocompatible hydrogel able to support the in vitro and in vivo viability and activity of hASC as well as the regeneration of osteochondral defects in dogs when implanted alone or with ASC.
Collapse
Affiliation(s)
- Cécile Boyer
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Gildas Réthoré
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- CHU Nantes, Service d’Odontologie Restauratrice et Chirurgicale, PHU4 OTONN, Nantes, France
| | - Pierre Weiss
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- CHU Nantes, Service d’Odontologie Restauratrice et Chirurgicale, PHU4 OTONN, Nantes, France
| | - Cyril d’Arros
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Julie Lesoeur
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- SC3M – “Electron Microscopy, Microcharacterization and Functional Morphohistology Imaging” Core Facility, Structure Fédérative de Recherche Franc̨ois Bonamy, INSERM – UMS016, CNRS 3556, CHU Nantes, Université de Nantes, Nantes, France
| | - Claire Vinatier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- SC3M – “Electron Microscopy, Microcharacterization and Functional Morphohistology Imaging” Core Facility, Structure Fédérative de Recherche Franc̨ois Bonamy, INSERM – UMS016, CNRS 3556, CHU Nantes, Université de Nantes, Nantes, France
| | - Boris Halgand
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- CHU Nantes, PHU4 OTONN, Nantes, France
| | - Olivier Geffroy
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Marion Fusellier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Gildas Vaillant
- CHU Nantes, PHU4 OTONN, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Patrice Roy
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Olivier Gauthier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- Centre of Research and Preclinical Investigation (C.R.I.P.), ONIRIS, Nantes, France
| | - Jérôme Guicheux
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
- SC3M – “Electron Microscopy, Microcharacterization and Functional Morphohistology Imaging” Core Facility, Structure Fédérative de Recherche Franc̨ois Bonamy, INSERM – UMS016, CNRS 3556, CHU Nantes, Université de Nantes, Nantes, France
- CHU Nantes, PHU4 OTONN, Nantes, France
| |
Collapse
|
12
|
Zhang J, Xu Y, Liu T, Min J, Ma Y, Song Y, Lu J, Mi W, Wang Y, Li H, Li W, Zhao D. In vivo construction of lymphoid node by implantation of adipose-derived stromal cells with hydroxypropyl methyl cellulose hydrogel in BALB/c nude mice. Organogenesis 2019; 15:85-99. [PMID: 31448695 DOI: 10.1080/15476278.2019.1656994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adipose-derived stromal cells have multilineage potential to differentiate into several specialized tissue types. Herein, we investigated whether ADSCs could differentiate into lymphoid node in vivo. Human ADSCs from routine liposuction were cultured in differentiation medium and were supplemented with transforming growth factor β1 (TGF)-β1 and basic fibroblast growth factor (bFGF). The induced hADSCs mixed with 13% (w/v) hydroxypropyl methylcellulose (HPMC) were injected into BALB/c nude mice subcutaneously. Eight weeks later, nodules were found under the injected sites. Histology, immunohistochemistry, and species identification analysis confirmed that the nodules were lymphoid nodes that were derived from the injected hADSCs. Our experiment demonstrated that the hADSCs could differentiate into lymphocyte-like cells and form lymphoid nodes in vivo. TGF-β1 and bFGF might play important roles in the differentiation of hADSCs into lymphocyte-like cells. Our study might present an alternative approach for engineering immune organs and thus offer potential treatment for immunodeficiency diseases.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Otolaryngology Head and Neck Surgery, the second affiliated hospital.,Department of Nephrology and Endocrinology, No.371 Central Hospital of People's Liberation Army , Xinxiang , Henan , People's Republic of China
| | - Yuqiao Xu
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Tao Liu
- Faculty of Biomedical Engineering
| | - Jie Min
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Yu Ma
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Yongli Song
- Department of Otolaryngology Head and Neck Surgery, the first affiliated hospital , Xi'an , China
| | - Jianrong Lu
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Wenjuan Mi
- Department of Otolaryngology Head and Neck Surgery, the first affiliated hospital , Xi'an , China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Hang Li
- State Key Laboratory of Cancer Biology and Department of Pathology, the first affiliated hospital , Xi'an , China
| | - Wangzhou Li
- Department of Plastic and Burns, the second affiliated hospital; Air Force Medical University , Xi'an , PR China
| | - Daqing Zhao
- Department of Otolaryngology Head and Neck Surgery, the second affiliated hospital
| |
Collapse
|
13
|
Perrier-Groult E, Pérès E, Pasdeloup M, Gazzolo L, Duc Dodon M, Mallein-Gerin F. Evaluation of the biocompatibility and stability of allogeneic tissue-engineered cartilage in humanized mice. PLoS One 2019; 14:e0217183. [PMID: 31107916 PMCID: PMC6527235 DOI: 10.1371/journal.pone.0217183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/07/2019] [Indexed: 11/18/2022] Open
Abstract
Articular cartilage (AC) has poor capacities of regeneration and lesions often lead to osteoarthritis. Current AC reconstruction implies autologous chondrocyte implantation which requires tissue sampling and grafting. An alternative approach would be to use scaffolds containing off-the-shelf allogeneic human articular chondrocytes (HACs). To investigate tolerance of allogeneic HACs by the human immune system, we developed a humanized mouse model implanted with allogeneic cartilage constructs generated in vitro. A prerequisite of the study was to identify a scaffold that would not provoke inflammatory reaction in host. Therefore, we first compared the response of hu-mice to two biomaterials used in regenerative medicine, collagen sponge and agarose hydrogel. Four weeks after implantation in hu-mice, acellular collagen sponges, but not acellular agarose hydrogels, showed positive staining for CD3 (T lymphocytes) and CD68 (macrophages), suggesting that collagen scaffold elicits weak inflammatory reaction. These data led us to deepen our evaluation of the biocompatibility of allogeneic tissue-engineered cartilage by using agarose as scaffold. Agarose hydrogels were combined with allogeneic HACs to reconstruct cartilage in vitro. Particular attention was paid to HLA-A2 compatibility between HACs to be grafted and immune human cells of hu-mice: HLA-A2+ or HLA-A2- HACs agarose hydrogels were cultured in the presence of a chondrogenic cocktail and implanted in HLA-A2+ hu-mice. After four weeks implantation and regardless of the HLA-A2 phenotype, chondrocytes were well-differentiated and produced cartilage matrix in agarose. In addition, no sign of T-cell or macrophage infiltration was seen in the cartilaginous constructs and no significant increase in subpopulations of T lymphocytes and monocytes was detected in peripheral blood and spleen. We show for the first time that humanized mouse represents a useful model to investigate human immune responsiveness to tissue-engineered cartilage and our data together indicate that allogeneic cartilage constructs can be suitable for cartilage engineering.
Collapse
Affiliation(s)
- Emeline Perrier-Groult
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
- * E-mail:
| | - Eléonore Pérès
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Marielle Pasdeloup
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
| | - Louis Gazzolo
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Madeleine Duc Dodon
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
| |
Collapse
|
14
|
Tourné-Péteilh C, Robin B, Lions M, Martinez J, Mehdi A, Subra G, Devoisselle JM. Combining sol–gel and microfluidics processes for the synthesis of protein-containing hybrid microgels. Chem Commun (Camb) 2019; 55:13112-13115. [DOI: 10.1039/c9cc04963k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biocompatible encapsulation of proteins in hybrid microgels of a silylated hydrogel, focused on soft procedures and cross-linking conditions.
Collapse
Affiliation(s)
| | | | | | | | - Ahmad Mehdi
- ICGM
- University of Montpellier
- CNRS
- ENSCM
- Montpellier
| | - Gilles Subra
- IBMM
- University of Montpellier
- CNRS
- ENSCM
- Montpellier
| | | |
Collapse
|
15
|
Cellulose-Based Superabsorbent Hydrogels. POLYMERS AND POLYMERIC COMPOSITES: A REFERENCE SERIES 2019. [DOI: 10.1007/978-3-319-77830-3_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Ribeiro AM, Magalhães M, Veiga F, Figueiras A. Cellulose-Based Hydrogels in Topical Drug Delivery: A Challenge in Medical Devices. POLYMERS AND POLYMERIC COMPOSITES: A REFERENCE SERIES 2019. [DOI: 10.1007/978-3-319-77830-3_41] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
17
|
|
18
|
Graceffa V, Vinatier C, Guicheux J, Stoddart M, Alini M, Zeugolis DI. Chasing Chimeras - The elusive stable chondrogenic phenotype. Biomaterials 2018; 192:199-225. [PMID: 30453216 DOI: 10.1016/j.biomaterials.2018.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
Abstract
The choice of the best-suited cell population for the regeneration of damaged or diseased cartilage depends on the effectiveness of culture conditions (e.g. media supplements, three-dimensional scaffolds, mechanical stimulation, oxygen tension, co-culture systems) to induce stable chondrogenic phenotype. Herein, advances and shortfalls in in vitro, preclinical and clinical setting of various in vitro microenvironment modulators on maintaining chondrocyte phenotype or directing stem cells towards chondrogenic lineage are critically discussed. Chondrocytes possess low isolation efficiency, limited proliferative potential and rapid phenotypic drift in culture. Mesenchymal stem cells are relatively readily available, possess high proliferation potential, exhibit great chondrogenic differentiation capacity, but they tend to acquire a hypertrophic phenotype when exposed to chondrogenic stimuli. Embryonic and induced pluripotent stem cells, despite their promising in vitro and preclinical data, are still under-investigated. Although a stable chondrogenic phenotype remains elusive, recent advances in in vitro microenvironment modulators are likely to develop clinically- and commercially-relevant therapies in the years to come.
Collapse
Affiliation(s)
- Valeria Graceffa
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Claire Vinatier
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Jerome Guicheux
- INSERMU1229, Regenerative Medicine and Skeleton (RMeS), University of Nantes, UFR Odontologie & CHU Nantes, PHU 4 OTONN, 44042 Nantes, France
| | - Martin Stoddart
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland.
| |
Collapse
|
19
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Injectable hydrogels: a new paradigm for osteochondral tissue engineering. J Mater Chem B 2018; 6:5499-5529. [PMID: 32254962 DOI: 10.1039/c8tb01430b] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteochondral tissue engineering has become a promising strategy for repairing focal chondral lesions and early osteoarthritis (OA), which account for progressive joint pain and disability in millions of people worldwide. Towards improving osteochondral tissue repair, injectable hydrogels have emerged as promising matrices due to their wider range of properties such as their high water content and porous framework, similarity to the natural extracellular matrix (ECM), ability to encapsulate cells within the matrix and ability to provide biological cues for cellular differentiation. Further, their properties such as those that facilitate minimally invasive deployment or delivery, and their ability to repair geometrically complex irregular defects have been critical for their success. In this review, we provide an overview of innovative approaches to engineer injectable hydrogels towards improved osteochondral tissue repair. Herein, we focus on understanding the biology of osteochondral tissue and osteoarthritis along with the need for injectable hydrogels in osteochondral tissue engineering. Furthermore, we discuss in detail different biomaterials (natural and synthetic) and various advanced fabrication methods being employed for the development of injectable hydrogels in osteochondral repair. In addition, in vitro and in vivo applications of developed injectable hydrogels for osteochondral tissue engineering are also reviewed. Finally, conclusions and future perspectives of using injectable hydrogels in osteochondral tissue engineering are provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India.
| | | | | | | |
Collapse
|
20
|
Adamski M, Fontana G, Gershlak JR, Gaudette GR, Le HD, Murphy WL. Two Methods for Decellularization of Plant Tissues for Tissue Engineering Applications. J Vis Exp 2018:57586. [PMID: 29912197 PMCID: PMC6101437 DOI: 10.3791/57586] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The autologous, synthetic, and animal-derived grafts currently used as scaffolds for tissue replacement have limitations due to low availability, poor biocompatibility, and cost. Plant tissues have favorable characteristics that make them uniquely suited for use as scaffolds, such as high surface area, excellent water transport and retention, interconnected porosity, preexisting vascular networks, and a wide range of mechanical properties. Two successful methods of plant decellularization for tissue engineering applications are described here. The first method is based on detergent baths to remove cellular matter, which is similar to previously established methods used to clear mammalian tissues. The second is a detergent-free method adapted from a protocol that isolates leaf vasculature and involves the use of a heated bleach and salt bath to clear the leaves and stems. Both methods yield scaffolds with comparable mechanical properties and low cellular metabolic impact, thus allowing the user to select the protocol which better suits their intended application.
Collapse
Affiliation(s)
| | - Gianluca Fontana
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health
| | - Joshua R Gershlak
- Department of Biomedical Engineering, Worcester Polytechnic Institute
| | - Glenn R Gaudette
- Department of Biomedical Engineering, Worcester Polytechnic Institute
| | - Hau D Le
- Department of Surgery, University of Wisconsin-Madison
| | - William L Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health; Department of Biomedical Engineering, University of Wisconsin College of Engineering;
| |
Collapse
|
21
|
Wong CC, Chen CH, Chiu LH, Tsuang YH, Bai MY, Chung RJ, Lin YH, Hsieh FJ, Chen YT, Yang TL. Facilitating In Vivo Articular Cartilage Repair by Tissue-Engineered Cartilage Grafts Produced From Auricular Chondrocytes. Am J Sports Med 2018; 46:713-727. [PMID: 29211970 DOI: 10.1177/0363546517741306] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Insufficient cell numbers still present a challenge for articular cartilage repair. Converting heterotopic auricular chondrocytes by extracellular matrix may be the solution. HYPOTHESIS Specific extracellular matrix may convert the phenotype of auricular chondrocytes toward articular cartilage for repair. STUDY DESIGN Controlled laboratory study. METHODS For in vitro study, rabbit auricular chondrocytes were cultured in monolayer for several passages until reaching status of dedifferentiation. Later, they were transferred to chondrogenic type II collagen (Col II)-coated plates for further cell conversion. Articular chondrogenic profiles, such as glycosaminoglycan deposition, articular chondrogenic gene, and protein expression, were evaluated after 14-day cultivation. Furthermore, 3-dimensional constructs were fabricated using Col II hydrogel-associated auricular chondrocytes, and their histological and biomechanical properties were analyzed. For in vivo study, focal osteochondral defects were created in the rabbit knee joints, and auricular Col II constructs were implanted for repair. RESULTS The auricular chondrocytes converted by a 2-step protocol expressed specific profiles of chondrogenic molecules associated with articular chondrocytes. The histological and biomechanical features of converted auricular chondrocytes became similar to those of articular chondrocytes when cultivated with Col II 3-dimensional scaffolds. In an in vivo animal model of osteochondral defects, the treated group (auricular Col II) showed better cartilage repair than did the control groups (sham, auricular cells, and Col II). Histological analyses revealed that cartilage repair was achieved in the treated groups with abundant type II collagen and glycosaminoglycans syntheses rather than elastin expression. CONCLUSION The study confirmed the feasibility of applying heterotopic chondrocytes for cartilage repair via extracellular matrix-induced cell conversion. CLINICAL RELEVANCE This study proposes a feasible methodology to convert heterotopic auricular chondrocytes for articular cartilage repair, which may serve as potential alternative sources for cartilage repair.
Collapse
Affiliation(s)
- Chin-Chean Wong
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hwa Chen
- Bone and Joint Research Center, Department of Orthopedics, Taipei Medical University Hospital, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Li-Hsuan Chiu
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, USA.,Center for Nano Tissue Engineering and Image Research, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yang-Hwei Tsuang
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Yi Bai
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan.,Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Ren-Jei Chung
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan
| | - Yun-Ho Lin
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Fon-Jou Hsieh
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - You-Tzung Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Lin Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.,a Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
22
|
Boyer C, Figueiredo L, Pace R, Lesoeur J, Rouillon T, Visage CL, Tassin JF, Weiss P, Guicheux J, Rethore G. Laponite nanoparticle-associated silated hydroxypropylmethyl cellulose as an injectable reinforced interpenetrating network hydrogel for cartilage tissue engineering. Acta Biomater 2018; 65:112-122. [PMID: 29128532 DOI: 10.1016/j.actbio.2017.11.027] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/20/2017] [Accepted: 11/07/2017] [Indexed: 11/26/2022]
Abstract
Articular cartilage is a connective tissue which does not spontaneously heal. To address this issue, biomaterial-assisted cell therapy has been researched with promising advances. The lack of strong mechanical properties is still a concern despite significant progress in three-dimensional scaffolds. This article's objective was to develop a composite hydrogel using a small amount of nano-reinforcement clay known as laponites. These laponites were capable of self-setting within the gel structure of the silated hydroxypropylmethyl cellulose (Si-HPMC) hydrogel. Laponites (XLG) were mixed with Si-HPMC to prepare composite hydrogels leading to the development of a hybrid interpenetrating network. This interpenetrating network increases the mechanical properties of the hydrogel. The in vitro investigations showed no side effects from the XLG regarding cytocompatibility or oxygen diffusion within the composite after cross-linking. The ability of the hybrid scaffold containing the composite hydrogel and chondrogenic cells to form a cartilaginous tissue in vivo was investigated during a 6-week implantation in subcutaneous pockets of nude mice. Histological analysis of the composite constructs revealed the formation of a cartilage-like tissue with an extracellular matrix containing glycosaminoglycans and collagens. Overall, this new hybrid construct demonstrates an interpenetrating network which enhances the hydrogel mechanical properties without interfering with its cytocompatibility, oxygen diffusion, or the ability of chondrogenic cells to self-organize in the cluster and produce extracellular matrix components. This composite hydrogel may be of relevance for the treatment of cartilage defects in a large animal model of articular cartilage defects. STATEMENT OF SIGNIFICANCE Articular cartilage is a tissue that fails to heal spontaneously. To address this clinically relevant issue, biomaterial-assisted cell therapy is considered promising but often lacks adequate mechanical properties. Our objective was to develop a composite hydrogel using a small amount of nano reinforcement (laponite) capable of gelling within polysaccharide based self-crosslinking hydrogel. This new hybrid construct demonstrates an interpenetrating network (IPN) which enhances the hydrogel mechanical properties without interfering with its cytocompatibility, O2 diffusion and the ability of chondrogenic cells to self-organize in cluster and produce extracellular matrix components. This composite hydrogel may be of relevance for the treatment of cartilage defects and will now be considered in a large animal model of articular cartilage defects.
Collapse
|
23
|
Calabrese G, Gulino R, Giuffrida R, Forte S, Figallo E, Fabbi C, Salvatorelli L, Memeo L, Gulisano M, Parenti R. In Vivo Evaluation of Biocompatibility and Chondrogenic Potential of a Cell-Free Collagen-Based Scaffold. Front Physiol 2017; 8:984. [PMID: 29238307 PMCID: PMC5712864 DOI: 10.3389/fphys.2017.00984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/17/2017] [Indexed: 01/07/2023] Open
Abstract
Injured articular cartilage has a limited innate regenerative capacity, due to the avascular nature and low cellularity of the tissue itself. Although several approaches have been proposed to repair the joint cartilage, none of them has proven to be effective. The absence of suitable therapeutic options has encouraged tissue-engineering approaches combining specific cell types and biomaterials. In the present work, we have evaluated the potential of a cell-free Collagen I-based scaffold to promote the augmentation of cartilage-like phenotype after subcutaneous implantation in the mouse. Forty female mice were grafted subcutaneously with scaffolds, while four additional mice without scaffold were used as negative controls. The effects of scaffold were evaluated at 1, 2, 4, 8, or 16 weeks after implantation. Immunohistochemical analysis shows the expression of typical cartilage markers, including type-II Collagen, Aggrecan, Matrilin-1 and Sox 9. These data are also confirmed by qRT-PCR that further show that both COL2A1 and COL1A1 increase over time, but the first one increases more rapidly, thus suggesting a typical cartilage-like address. Histological analysis shows the presence of some pericellular lacunae, after 8 and 16 weeks. Results suggest that this scaffold (i) is biocompatible in vivo, (ii) is able to recruit host cells (iii) induce chondrogenic differentiation of host cells. Such evidences suggest that this cell-free scaffold is promising and represents a potential approach for cartilage regeneration.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Istituto Oncologico del Mediterraneo Ricerca, Catania, Italy
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Istituto Oncologico del Mediterraneo Ricerca, Catania, Italy
| | | | - Stefano Forte
- Istituto Oncologico del Mediterraneo Ricerca, Catania, Italy
| | | | | | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, "Policlinico Vittorio Emanuele", Anatomic Pathology Section, University of Catania, Catania, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Italy
| | - Massimo Gulisano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
24
|
Henry N, Clouet J, Fragale A, Griveau L, Chédeville C, Véziers J, Weiss P, Le Bideau J, Guicheux J, Le Visage C. Pullulan microbeads/Si-HPMC hydrogel injectable system for the sustained delivery of GDF-5 and TGF-β1: new insight into intervertebral disc regenerative medicine. Drug Deliv 2017; 24:999-1010. [PMID: 28645219 PMCID: PMC8241148 DOI: 10.1080/10717544.2017.1340362] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/31/2017] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Discogenic low back pain is considered a major health concern and no etiological treatments are today available to tackle this disease. To clinically address this issue at early stages, there is a rising interest in the stimulation of local cells by in situ injection of growth factors targeting intervertebral disc (IVD) degenerative process. Despite encouraging safety and tolerability results in clinic, growth factors efficacy may be further improved. To this end, the use of a delivery system allowing a sustained release, while protecting growth factors from degradation appears of particular interest. We propose herein the design of a new injectable biphasic system, based on the association of pullulan microbeads (PMBs) into a cellulose-based hydrogel (Si-HPMC), for the TGF-β1 and GDF-5 growth factors sustained delivery. We present for the first time the design and mechanical characterization of both the PMBs and the called biphasic system (PMBs/Si-HPMC). Their loading and release capacities were also studied and we were able to demonstrate a sustained release of both growth factors, for up to 28 days. Noteworthy, the growth factors biological activity on human cells was maintained. Altogether, these data suggest that this PMBs/Si-HPMC biphasic system may be a promising candidate for the development of an innovative bioactive delivery system for IVD regenerative medicine.
Collapse
Affiliation(s)
- Nina Henry
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- Institut des Matériaux Jean Rouxel (IMN), Université de Nantes, CNRS, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
| | - Johann Clouet
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- CHU Nantes, PHU 11 Pharmacie, Pharmacie Centrale, Nantes, France
- UFR Sciences Biologiques et Pharmaceutiques, Université de Nantes, Nantes, France
| | - Audrey Fragale
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
| | - Louise Griveau
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
| | - Claire Chédeville
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
| | - Joëlle Véziers
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- SC3M platform, UMS INSERM 016/CNRS 3556, SFR François Bonamy, Nantes, France
- CHU Nantes, PHU 4 OTONN, Nantes, France
| | - Pierre Weiss
- UFR Odontologie, Université de Nantes, Nantes, France
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team REGOS “Regenerative Medicine of Bone Tissues”, Nantes, France
| | - Jean Le Bideau
- Institut des Matériaux Jean Rouxel (IMN), Université de Nantes, CNRS, Nantes, France
| | - Jérôme Guicheux
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- CHU Nantes, PHU 4 OTONN, Nantes, France
| | - Catherine Le Visage
- INSERM, UMRS 1229, RMeS “Regenerative Medicine and Skeleton”, Team STEP “Skeletal Physiopathology and Joint Regenerative Medicine”, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
| |
Collapse
|
25
|
Microgels of silylated HPMC as a multimodal system for drug co-encapsulation. Int J Pharm 2017; 532:790-801. [PMID: 28755992 DOI: 10.1016/j.ijpharm.2017.07.074] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 01/22/2023]
Abstract
Combined therapy is a global strategy developed to prevent drug resistance in cancer and infectious diseases. In this field, there is a need of multifunctional drug delivery systems able to co-encapsulate small drug molecules, peptides, proteins, associated to targeting functions, nanoparticles. Silylated hydrogels are alkoxysilane hybrid polymers that can be engaged in a sol-gel process, providing chemical cross linking in physiological conditions, and functionalized biocompatible hybrid materials. In the present work, microgels were prepared with silylated (hydroxypropyl)methyl cellulose (Si-HPMC) that was chemically cross linked in soft conditions of pH and temperature. They were prepared by an emulsion templating process, water in oil (W/O), as microreactors where the condensation reaction took place. The ability to functionalize the microgels, so-called FMGs, in a one-pot process, was evaluated by grafting a silylated hydrophilic model drug, fluorescein (Si-Fluor), using the same reaction of condensation. Biphasic microgels (BPMGs) were prepared to evaluate their potential to encapsulate lipophilic model drug (Nile red). They were composed of two separate compartments, one oily phase (sesame oil) trapped in the cross linked Si-HPMC hydrophilic phase. The FMGs and BPMGs were characterized by different microscopic techniques (optic, epi-fluorescence, Confocal Laser Scanning Microscopy and scanning electronic microscopy), the mechanical properties were monitored using nano indentation by Atomic Force Microscopy (AFM), and different preliminary tests were performed to evaluate their chemical and physical stability. Finally, it was demonstrated that it is possible to co-encapsulate both hydrophilic and hydrophobic drugs, in silylated microgels, that were physically and chemically stable. They were obtained by chemical cross linking in soft conditions, and without surfactant addition during the emulsification process. The amount of drug loaded was in favor of further biological activity. Mechanical stimulations should be necessary to trigger drug release.
Collapse
|
26
|
Bioreactor mechanically guided 3D mesenchymal stem cell chondrogenesis using a biocompatible novel thermo-reversible methylcellulose-based hydrogel. Sci Rep 2017; 7:45018. [PMID: 28332587 PMCID: PMC5362895 DOI: 10.1038/srep45018] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/17/2017] [Indexed: 12/14/2022] Open
Abstract
Autologous chondrocyte implantation for cartilage repair represents a challenge because strongly limited by chondrocytes' poor expansion capacity in vitro. Mesenchymal stem cells (MSCs) can differentiate into chondrocytes, while mechanical loading has been proposed as alternative strategy to induce chondrogenesis excluding the use of exogenous factors. Moreover, MSC supporting material selection is fundamental to allow for an active interaction with cells. Here, we tested a novel thermo-reversible hydrogel composed of 8% w/v methylcellulose (MC) in a 0.05 M Na2SO4 solution. MC hydrogel was obtained by dispersion technique and its thermo-reversibility, mechanical properties, degradation and swelling were investigated, demonstrating a solution-gelation transition between 34 and 37 °C and a low bulk degradation (<20%) after 1 month. The lack of any hydrogel-derived immunoreaction was demonstrated in vivo by mice subcutaneous implantation. To induce in vitro chondrogenesis, MSCs were seeded into MC solution retained within a porous polyurethane (PU) matrix. PU-MC composites were subjected to a combination of compression and shear forces for 21 days in a custom made bioreactor. Mechanical stimulation led to a significant increase in chondrogenic gene expression, while histological analysis detected sulphated glycosaminoglycans and collagen II only in loaded specimens, confirming MC hydrogel suitability to support load induced MSCs chondrogenesis.
Collapse
|
27
|
Calabrese G, Forte S, Gulino R, Cefalì F, Figallo E, Salvatorelli L, Maniscalchi ET, Angelico G, Parenti R, Gulisano M, Memeo L, Giuffrida R. Combination of Collagen-Based Scaffold and Bioactive Factors Induces Adipose-Derived Mesenchymal Stem Cells Chondrogenic Differentiation In vitro. Front Physiol 2017; 8:50. [PMID: 28210226 PMCID: PMC5288372 DOI: 10.3389/fphys.2017.00050] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/18/2017] [Indexed: 12/27/2022] Open
Abstract
Recently, multipotent mesenchymal stem cells (MSCs) have attracted much attention in the field of regenerative medicine due to their ability to give rise to different cell types, including chondrocytes. Damaged articular cartilage repair is one of the most challenging issues for regenerative medicine, due to the intrinsic limited capability of cartilage to heal because of its avascular nature. While surgical approaches like chondral autografts and allografts provide symptoms and function improvement only for a short period, MSC based stimulation therapies, like microfracture surgery or autologous matrix-induced chondrogenesis demonstrate to be more effective. The use of adult chondrocytes, which are the main cellular constituent of cartilage, in medical practice, is indeed limited due to their instability in monolayer culture and difficulty to collect donor tissue (articular and nasal cartilage). The most recent cartilage engineering approaches combine cells, biomaterial scaffold and bioactive factors to promote functional tissue replacements. Many recent evidences demonstrate that scaffolds providing specific microenvironmental conditions can promote MSCs differentiation toward a functional phenotype. In the present work, the chondrogenic potential of a new Collagen I based 3D scaffold has been assessed in vitro, in combination with human adipose-derived MSCs which possess a higher chondrogenic potential compared to MSCs isolated from other tissues. Our data indicate that the scaffold was able to promote the early stages of chondrogenic commitment and that supplementation of specific soluble factors was able to induce the complete differentiation of MSCs in chondrocytes as demonstrated by the appearance of cartilage distinctive markers (Sox 9, Aggrecan, Matrilin-1, and Collagen II), as well as by the cartilage-specific Alcian Blue staining and by the acquisition of typical cellular morphology. Such evidences suggest that the investigated scaffold formulation could be suitable for the production of medical devices that can be beneficial in the field of articular cartilage engineering, thus improving the efficacy and durability of the current therapeutic options.
Collapse
Affiliation(s)
- Giovanna Calabrese
- Istituto Oncologico del Mediterraneo - Ricerca ViagrandeCatania, Italy; Physiology Section, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Stefano Forte
- Istituto Oncologico del Mediterraneo - Ricerca Viagrande Catania, Italy
| | - Rosario Gulino
- Istituto Oncologico del Mediterraneo - Ricerca ViagrandeCatania, Italy; Physiology Section, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | | | | | - Lucia Salvatorelli
- Anatomic Pathology Section, Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, "Policlinico Vittorio Emanuele", University of Catania Catania, Italy
| | - Eugenia T Maniscalchi
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Giuseppe Angelico
- Anatomic Pathology Section, Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, "Policlinico Vittorio Emanuele", University of Catania Catania, Italy
| | - Rosalba Parenti
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Massimo Gulisano
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology Viagrande, Italy
| | | |
Collapse
|
28
|
Zhang J, Liu W, Gauthier O, Sourice S, Pilet P, Rethore G, Khairoun K, Bouler JM, Tancret F, Weiss P. A simple and effective approach to prepare injectable macroporous calcium phosphate cement for bone repair: Syringe-foaming using a viscous hydrophilic polymeric solution. Acta Biomater 2016; 31:326-338. [PMID: 26631875 DOI: 10.1016/j.actbio.2015.11.055] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/28/2015] [Accepted: 11/25/2015] [Indexed: 11/18/2022]
Abstract
In this study, we propose a simple and effective strategy to prepare injectable macroporous calcium phosphate cements (CPCs) by syringe-foaming via hydrophilic viscous polymeric solution, such as using silanized-hydroxypropyl methylcellulose (Si-HPMC) as a foaming agent. The Si-HPMC foamed CPCs demonstrate excellent handling properties such as injectability and cohesion. After hardening the foamed CPCs possess hierarchical macropores and their mechanical properties (Young's modulus and compressive strength) are comparable to those of cancellous bone. Moreover, a preliminary in vivo study in the distal femoral sites of rabbits was conducted to evaluate the biofunctionality of this injectable macroporous CPC. The evidence of newly formed bone in the central zone of implantation site indicates the feasibility and effectiveness of this foaming strategy that will have to be optimized by further extensive animal experiments. STATEMENT OF SIGNIFICANCE A major challenge in the design of biomaterial-based injectable bone substitutes is the development of cohesive, macroporous and self-setting calcium phosphate cement (CPC) that enables rapid cell invasion with adequate initial mechanical properties without the use of complex processing and additives. Thus, we propose a simple and effective strategy to prepare injectable macroporous CPCs through syringe-foaming using a hydrophilic viscous polymeric solution (silanized-hydroxypropyl methylcellulose, Si-HPMC) as a foaming agent, that simultaneously meets all the aforementioned aims. Evidence from our in vivo studies shows the existence of newly formed bone within the implantation site, indicating the feasibility and effectiveness of this foaming strategy, which could be used in various CPC systems using other hydrophilic viscous polymeric solutions.
Collapse
Affiliation(s)
- Jingtao Zhang
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France; Université de Nantes, Polytech Nantes, Institut des Matériaux Jean Rouxel, CNRS UMR 6502, Rue Christian Pauc, BP 50609, 44306 Nantes Cedex 3, France
| | - Weizhen Liu
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France; Université de Nantes, Polytech Nantes, Institut des Matériaux Jean Rouxel, CNRS UMR 6502, Rue Christian Pauc, BP 50609, 44306 Nantes Cedex 3, France
| | - Olivier Gauthier
- ONIRIS - Ecole Nationale Veterinaire de Nantes, Atlanpole-La Chantrerie, BP 40706, 44307 Nantes cedex 3, France
| | - Sophie Sourice
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France
| | - Paul Pilet
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France; CHU de Nantes, Nantes University Hospital, PHU 4 OTONN, 1 Pl A. Ricordeau Nantes, France
| | - Gildas Rethore
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France; CHU de Nantes, Nantes University Hospital, PHU 4 OTONN, 1 Pl A. Ricordeau Nantes, France
| | - Khalid Khairoun
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France
| | - Jean-Michel Bouler
- Université de Nantes, CEISAM, CNRS UMR 6230, 2 rue de la Houssinière, BP 92208, 44322 Nantes Cedex 3, France
| | - Franck Tancret
- Université de Nantes, Polytech Nantes, Institut des Matériaux Jean Rouxel, CNRS UMR 6502, Rue Christian Pauc, BP 50609, 44306 Nantes Cedex 3, France
| | - Pierre Weiss
- Université de Nantes, INSERM UMRS 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, 1 place Alexis Ricordeau, BP 84215, 44042 Nantes Cedex 1, France; CHU de Nantes, Nantes University Hospital, PHU 4 OTONN, 1 Pl A. Ricordeau Nantes, France.
| |
Collapse
|
29
|
Rederstorff E, Rethore G, Weiss P, Sourice S, Beck-Cormier S, Mathieu E, Maillasson M, Jacques Y, Colliec-Jouault S, Fellah BH, Guicheux J, Vinatier C. Enriching a cellulose hydrogel with a biologically active marine exopolysaccharide for cell-based cartilage engineering. J Tissue Eng Regen Med 2015; 11:1152-1164. [PMID: 25824373 DOI: 10.1002/term.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/22/2014] [Accepted: 01/15/2015] [Indexed: 11/09/2022]
Abstract
The development of biologically and mechanically competent hydrogels is a prerequisite in cartilage engineering. We recently demonstrated that a marine exopolysaccharide, GY785, stimulates the in vitro chondrogenesis of adipose stromal cells. In the present study, we thus hypothesized that enriching our silated hydroxypropyl methylcellulose hydrogel (Si-HPMC) with GY785 might offer new prospects in the development of scaffolds for cartilage regeneration. The interaction properties of GY785 with growth factors was tested by surface plasmon resonance (SPR). The biocompatibility of Si-HPMC/GY785 towards rabbit articular chondrocytes (RACs) and its ability to maintain and recover a chondrocytic phenotype were then evaluated in vitro by MTS assay, cell counting and qRT-PCR. Finally, we evaluated the potential of Si-HPMC/GY785 associated with RACs to form cartilaginous tissue in vivo by transplantation into the subcutis of nude mice for 3 weeks. Our SPR data indicated that GY785 was able to physically interact with BMP-2 and TGFβ. Our analyses also showed that three-dimensionally (3D)-cultured RACs into Si-HPMC/GY785 strongly expressed type II collagen (COL2) and aggrecan transcripts when compared to Si-HPMC alone. In addition, RACs also produced large amounts of extracellular matrix (ECM) containing glycosaminoglycans (GAG) and COL2. When dedifferentiated RACs were replaced in 3D in Si-HPMC/GY785, the expressions of COL2 and aggrecan transcripts were recovered and that of type I collagen decreased. Immunohistological analyses of Si-HPMC/GY785 constructs transplanted into nude mice revealed the production of a cartilage-like extracellular matrix (ECM) containing high amounts of GAG and COL2. These results indicate that GY785-enriched Si-HPMC appears to be a promising hydrogel for cartilage tissue engineering. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- E Rederstorff
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France.,French Research Institute for Exploitation of the Sea (IFREMER), Laboratory of Biotechnology and Marine Molecules, Nantes, France
| | - G Rethore
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France.,Centre Hospitalier Universitaire Nantes, PHU4, Ostéo-articulaire Tête et Cou, Odontologie, Neurochirurgie, Neurotraumatologie (OTONN), Nantes, France
| | - P Weiss
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France.,Centre Hospitalier Universitaire Nantes, PHU4, Ostéo-articulaire Tête et Cou, Odontologie, Neurochirurgie, Neurotraumatologie (OTONN), Nantes, France
| | - S Sourice
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France
| | - S Beck-Cormier
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France
| | - E Mathieu
- INSERM, UMRS 1087, L'Institut du Thorax, Nantes, France
| | - M Maillasson
- INSERM, UMRS 1087, L'Institut du Thorax, Nantes, France.,Plateforme IMPACT Biogenouest, CRCNA-INSERM U892, SFR Santé François Bonamy/UMS INSERM, Nantes, France
| | - Y Jacques
- INSERM, UMRS 1087, L'Institut du Thorax, Nantes, France.,Plateforme IMPACT Biogenouest, CRCNA-INSERM U892, SFR Santé François Bonamy/UMS INSERM, Nantes, France
| | - S Colliec-Jouault
- French Research Institute for Exploitation of the Sea (IFREMER), Laboratory of Biotechnology and Marine Molecules, Nantes, France
| | - B H Fellah
- Centre for Preclinical Research and Investigation of the ONIRIS, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering (CRIP), Nantes, France
| | - J Guicheux
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France.,Centre Hospitalier Universitaire Nantes, PHU4, Ostéo-articulaire Tête et Cou, Odontologie, Neurochirurgie, Neurotraumatologie (OTONN), Nantes, France
| | - C Vinatier
- INSERM, UMRS 791-LIOAD, Skeletal Tissue Engineering and Physiopathology (STEP) Group, UFR Odontology, Nantes, France.,Université de Nantes, Unité de Formation et de Recherche (UFR) Odontologie, Nantes, France
| |
Collapse
|
30
|
Implantation of encapsulated human septal chondrocytes into immunocompetent mice using alginate microfibers. BIOCHIP JOURNAL 2015. [DOI: 10.1007/s13206-014-9109-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
31
|
Kawalec M, Sitkowska A, Sobota M, Sieroń AL, Komar P, Kurcok P. Human procollagen type I surface-modified PHB-based non-woven textile scaffolds for cell growth: preparation and short-term biological tests. Biomed Mater 2014; 9:065005. [DOI: 10.1088/1748-6041/9/6/065005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
32
|
Bhardwaj N, Devi D, Mandal BB. Tissue-engineered cartilage: the crossroads of biomaterials, cells and stimulating factors. Macromol Biosci 2014; 15:153-82. [PMID: 25283763 DOI: 10.1002/mabi.201400335] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/25/2014] [Indexed: 02/06/2023]
Abstract
Damage to cartilage represents one of the most challenging tasks of musculoskeletal therapeutics due to its limited propensity for healing and regenerative capabilities. Lack of current treatments to restore cartilage tissue function has prompted research in this rapidly emerging field of tissue regeneration of functional cartilage tissue substitutes. The development of cartilaginous tissue largely depends on the combination of appropriate biomaterials, cell source, and stimulating factors. Over the years, various biomaterials have been utilized for cartilage repair, but outcomes are far from achieving native cartilage architecture and function. This highlights the need for exploration of suitable biomaterials and stimulating factors for cartilage regeneration. With these perspectives, we aim to present an overview of cartilage tissue engineering with recent progress, development, and major steps taken toward the generation of functional cartilage tissue. In this review, we have discussed the advances and problems in tissue engineering of cartilage with strong emphasis on the utilization of natural polymeric biomaterials, various cell sources, and stimulating factors such as biophysical stimuli, mechanical stimuli, dynamic culture, and growth factors used so far in cartilage regeneration. Finally, we have focused on clinical trials, recent innovations, and future prospects related to cartilage engineering.
Collapse
Affiliation(s)
- Nandana Bhardwaj
- Seri-Biotechnology Unit, Life Science Division, Institute of Advanced Study in Science and Technology, Guwahati, 781035, India
| | | | | |
Collapse
|
33
|
Zeng L, Chen X, Zhang Q, Yu F, Li Y, Yao Y. Redifferentiation of dedifferentiated chondrocytes in a novel three‐dimensional microcavitary hydrogel. J Biomed Mater Res A 2014; 103:1693-702. [DOI: 10.1002/jbm.a.35309] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/31/2014] [Accepted: 08/06/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Lei Zeng
- School of Materials Science and Engineering, South China University of TechnologyGuangzhou510641 China
- National Engineering Research Center for Tissue Restoration and ReconstructionGuangzhou510006 China
| | - Xiaofeng Chen
- School of Materials Science and Engineering, South China University of TechnologyGuangzhou510641 China
- National Engineering Research Center for Tissue Restoration and ReconstructionGuangzhou510006 China
| | - Qing Zhang
- School of Materials Science and Engineering, South China University of TechnologyGuangzhou510641 China
- National Engineering Research Center for Tissue Restoration and ReconstructionGuangzhou510006 China
| | - Feng Yu
- School of Materials Science and Engineering, South China University of TechnologyGuangzhou510641 China
- National Engineering Research Center for Tissue Restoration and ReconstructionGuangzhou510006 China
| | - Yuli Li
- School of Materials Science and Engineering, South China University of TechnologyGuangzhou510641 China
- National Engineering Research Center for Tissue Restoration and ReconstructionGuangzhou510006 China
| | - Yongchang Yao
- School of Materials Science and Engineering, South China University of TechnologyGuangzhou510641 China
- National Engineering Research Center for Tissue Restoration and ReconstructionGuangzhou510006 China
| |
Collapse
|
34
|
Liu W, Zhang J, Rethore G, Khairoun K, Pilet P, Tancret F, Bouler JM, Weiss P. A novel injectable, cohesive and toughened Si-HPMC (silanized-hydroxypropyl methylcellulose) composite calcium phosphate cement for bone substitution. Acta Biomater 2014; 10:3335-45. [PMID: 24657196 DOI: 10.1016/j.actbio.2014.03.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/11/2014] [Accepted: 03/11/2014] [Indexed: 10/25/2022]
Abstract
This study reports on the incorporation of the self-setting polysaccharide derivative hydrogel (silanized-hydroxypropyl methylcellulose, Si-HPMC) into the formulation of calcium phosphate cements (CPCs) to develop a novel injectable material for bone substitution. The effects of Si-HPMC on the handling properties (injectability, cohesion and setting time) and mechanical properties (Young's modulus, fracture toughness, flexural and compressive strength) of CPCs were systematically studied. It was found that Si-HPMC could endow composite CPC pastes with an appealing rheological behavior at the early stage of setting, promoting its application in open bone cavities. Moreover, Si-HPMC gave the composite CPC good injectability and cohesion, and reduced the setting time. Si-HPMC increased the porosity of CPCs after hardening, especially the macroporosity as a result of entrapped air bubbles; however, it improved, rather than compromised, the mechanical properties of composite CPCs, which demonstrates a strong toughening and strengthening effect. In view of the above, the Si-HPMC composite CPC may be particularly promising as bone substitute material for clinic application.
Collapse
|
35
|
|
36
|
Abstract
Currently, the gold standard for reconstruction after rhinectomy or severe trauma to the nose, includes transposition of autologous mucosal flaps plus autologous cartilage grating and coverage using a skin flap. Difficulties with this approach arise where; cartilage and mucosa harvested from autologous donor sites is insufficient to achieve a passable aesthetic and functional reconstruction. Skin flaps are often bulky, poor color matches with hair follicles that reduce the aesthetic quality of the reconstruction. We suggest that tissue engineering could be a source of functional replacement tissues for nasal reconstructive surgery. However, the advancement of such an approach is dependent on the dissemination of scientific information into the clinical community, regarding the engineering of tissues such as mucosa, skin, and cartilage. This paper therefore reviews how the tissue engineering strategies available for producing clinically viable tissues could help resolve issues around reconstructing the human nose.
Collapse
|
37
|
Effect of microcavitary alginate hydrogel with different pore sizes on chondrocyte culture for cartilage tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 34:168-75. [DOI: 10.1016/j.msec.2013.09.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/31/2013] [Accepted: 09/06/2013] [Indexed: 01/28/2023]
|
38
|
Adipose stromal cells contain phenotypically distinct adipogenic progenitors derived from neural crest. PLoS One 2013; 8:e84206. [PMID: 24391913 PMCID: PMC3877257 DOI: 10.1371/journal.pone.0084206] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 11/13/2013] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that adipose-derived stromal/stem cells (ASCs) contain phenotypically and functionally heterogeneous subpopulations of cells, but their developmental origin and their relative differentiation potential remain elusive. In the present study, we aimed at investigating how and to what extent the neural crest contributes to ASCs using Cre-loxP-mediated fate mapping. ASCs harvested from subcutaneous fat depots of either adult P0-Cre/or Wnt1-Cre/Floxed-reporter mice contained a few neural crest-derived ASCs (NCDASCs). This subpopulation of cells was successfully expanded in vitro under standard culture conditions and their growth rate was comparable to non-neural crest derivatives. Although NCDASCs were positive for several mesenchymal stem cell markers as non-neural crest derivatives, they exhibited a unique bipolar or multipolar morphology with higher expression of markers for both neural crest progenitors (p75NTR, Nestin, and Sox2) and preadipocytes (CD24, CD34, S100, Pref-1, GATA2, and C/EBP-delta). NCDASCs were able to differentiate into adipocytes with high efficiency but their osteogenic and chondrogenic potential was markedly attenuated, indicating their commitment to adipogenesis. In vivo, a very small proportion of adipocytes were originated from the neural crest. In addition, p75NTR-positive neural crest-derived cells were identified along the vessels within the subcutaneous adipose tissue, but they were negative for mural and endothelial markers. These results demonstrate that ASCs contain neural crest-derived adipocyte-restricted progenitors whose phenotype is distinct from that of non-neural crest derivatives.
Collapse
|
39
|
The role of adipose-derived stromal cells and hydroxypropylmethylcellulose in engineering cartilage tissue in vivo. Cytotechnology 2013; 66:779-90. [PMID: 24287610 DOI: 10.1007/s10616-013-9627-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/26/2013] [Indexed: 12/25/2022] Open
Abstract
This study demonstrated a newly developed method using adipose tissue-derived stromal cells (ADSCs) and hydroxypropylmethylcellulose (HPMC) in building injectable tissue engineered cartilage in vivo. ADSCs from rabbit subcutaneous fatty tissue were cultured in chondrogenic differentiation medium and supplemented with transforming growth factor-β1 (TGF-β1) and basic fibroblast growth factor (bFGF). Histological, immunohistochemistry and RT-PCR analysis confirmed that the ADSCs differentiated into chondrocytes following induction. Induced ADSCs mixed with 15 % HPMC were injected into the subcutaneous tissue of nude mice and, after a period of 8 weeks, newly formed cartilage was observed at the site of injection. The ability of ADSCs cultured in the induction medium with TGF-β1 and bFGF to differentiate into chondrocytes and construct new cartilage indicates that ADSCs are suitable for use as seed cells in cartilage tissue engineering. HPMC, according to its good water solubility and being able to transform from liquid to solid at body temperature, was found to be an ideal scaffold for tissue engineering.
Collapse
|
40
|
Khan F, Ahmad SR. Biomimetic Polysaccharides and Derivatives for Cartilage Tissue Regeneration. Biomimetics (Basel) 2013. [DOI: 10.1002/9781118810408.ch1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
41
|
Lohan A, Marzahn U, El Sayed K, Bock C, Haisch A, Kohl B, Stoelzel K, John T, Ertel W, Schulze-Tanzil G. Heterotopic and orthotopic autologous chondrocyte implantation using a minipig chondral defect model. Ann Anat 2013; 195:488-97. [PMID: 23742980 DOI: 10.1016/j.aanat.2013.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 02/24/2013] [Accepted: 04/02/2013] [Indexed: 01/21/2023]
Abstract
Implantation of non-articular (heterotopic) chondrocyte-based implants might be an alternative approach to articular cartilage repair. This strategy could be helpful in cases in which there are no or too few articular chondrocytes available. Therefore, this study was undertaken to compare joint cartilage defect healing in the minipig model after implantation of heterotopic auricular and orthotopic articular chondrocytes. Poly-glycolic acid (PGA) associated three-dimensional (3D) constructs were prepared culturing autologous minipig-derived articular and auricular chondrocytes for 7 days in a dynamic culture system. Chondrocyte PGA constructs were implanted into 8mm diameter and ∼1.1mm deep chondral defects within the medial and lateral condyles of the minipig knee joints. Empty defects served as controls for assessment of the intrinsic healing response. Defect healing was monitored 6 months post implantation using a macroscopic and microscopic score system and biomechanical analysis. Neo-cartilage formation could be observed in the PGA constructs seeded with articular and auricular chondrocytes in vivo. The defect healing did not significantly differ at the macroscopic and histological level in response to implantation of either autologous articular or auricular chondrocytes seeded constructs compared with the empty defects. Although the differences were not significant, the auricular chondrocytes-based implants led to a slightly inferior repair quality at the macroscopic level, but a histologically superior healing response when compared with the empty defect group. However, biomechanical analysis revealed a higher stiffness in repair tissues produced by auricular chondrocyte implantation compared with the other groups. Deduced from these results, articular chondrocytes represent the preferable cell source for implantation.
Collapse
Affiliation(s)
- Anke Lohan
- Department for Orthopaedic, Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Effects of in vitro low oxygen tension preconditioning of adipose stromal cells on their in vivo chondrogenic potential: application in cartilage tissue repair. PLoS One 2013; 8:e62368. [PMID: 23638053 PMCID: PMC3640047 DOI: 10.1371/journal.pone.0062368] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 03/20/2013] [Indexed: 12/16/2022] Open
Abstract
Purpose Multipotent stromal cell (MSC)-based regenerative strategy has shown promise for the repair of cartilage, an avascular tissue in which cells experience hypoxia. Hypoxia is known to promote the early chondrogenic differentiation of MSC. The aim of our study was therefore to determine whether low oxygen tension could be used to enhance the regenerative potential of MSC for cartilage repair. Methods MSC from rabbit or human adipose stromal cells (ASC) were preconditioned in vitro in control or chondrogenic (ITS and TGF-β) medium and in 21 or 5% O2. Chondrogenic commitment was monitored by measuring COL2A1 and ACAN expression (real-time PCR). Preconditioned rabbit and human ASC were then incorporated into an Si-HPMC hydrogel and injected (i) into rabbit articular cartilage defects for 18 weeks or (ii) subcutaneously into nude mice for five weeks. The newly formed tissue was qualitatively and quantitatively evaluated by cartilage-specific immunohistological staining and scoring. The phenotype of ASC cultured in a monolayer or within Si-HPMC in control or chondrogenic medium and in 21 or 5% O2 was finally evaluated using real-time PCR. Results/Conclusions 5% O2 increased the in vitro expression of chondrogenic markers in ASC cultured in induction medium. Cells implanted within Si-HPMC hydrogel and preconditioned in chondrogenic medium formed a cartilaginous tissue, regardless of the level of oxygen. In addition, the 3D in vitro culture of ASC within Si-HPMC hydrogel was found to reinforce the pro-chondrogenic effects of the induction medium and 5% O2. These data together indicate that although 5% O2 enhances the in vitro chondrogenic differentiation of ASC, it does not enhance their in vivo chondrogenesis. These results also highlight the in vivo chondrogenic potential of ASC and their potential value in cartilage repair.
Collapse
|
43
|
Khan F, Ahmad SR. Polysaccharides and Their Derivatives for Versatile Tissue Engineering Application. Macromol Biosci 2013; 13:395-421. [DOI: 10.1002/mabi.201200409] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/06/2013] [Indexed: 12/13/2022]
|
44
|
Mathieu E, Lamirault G, Toquet C, Lhommet P, Rederstorff E, Sourice S, Biteau K, Hulin P, Forest V, Weiss P, Guicheux J, Lemarchand P. Intramyocardial delivery of mesenchymal stem cell-seeded hydrogel preserves cardiac function and attenuates ventricular remodeling after myocardial infarction. PLoS One 2012; 7:e51991. [PMID: 23284842 PMCID: PMC3527411 DOI: 10.1371/journal.pone.0051991] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 11/09/2012] [Indexed: 12/18/2022] Open
Abstract
Background To improve the efficacy of bone marrow-derived mesenchymal stem cell (MSC) therapy targeted to infarcted myocardium, we investigated whether a self-setting silanized hydroxypropyl methylcellulose (Si-HPMC) hydrogel seeded with MSC (MSC+hydrogel) could preserve cardiac function and attenuate left ventricular (LV) remodeling during an 8-week follow-up study in a rat model of myocardial infarction (MI). Methodology/Principal Finding Si-HPMC hydrogel alone, MSC alone or MSC+hydrogel were injected into the myocardium immediately after coronary artery ligation in female Lewis rats. Animals in the MSC+hydrogel group showed an increase in cardiac function up to 28 days after MI and a mid-term prevention of cardiac function alteration at day 56. Histological analyses indicated that the injection of MSC+hydrogel induced a decrease in MI size and an increase in scar thickness and ultimately limited the transmural extent of MI. These findings show that intramyocardial injection of MSC+hydrogel induced short-term recovery of ventricular function and mid-term attenuation of remodeling after MI. Conclusion/Significance These beneficial effects may be related to the specific scaffolding properties of the Si-HPMC hydrogel that may provide the ability to support MSC injection and engraftment within myocardium.
Collapse
Affiliation(s)
- Eva Mathieu
- INSERM UMR1087, CNRS UMR6291, l’institut du thorax, Nantes, France
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
| | - Guillaume Lamirault
- INSERM UMR1087, CNRS UMR6291, l’institut du thorax, Nantes, France
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- CHU de Nantes, Nantes, France
| | - Claire Toquet
- CHU de Nantes, Nantes, France
- Service d’Anatomie Pathologique, E.A. Biometadys, CHU de Nantes, Nantes, France
| | - Pierre Lhommet
- INSERM UMR1087, CNRS UMR6291, l’institut du thorax, Nantes, France
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
| | - Emilie Rederstorff
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- INSERM, U791, Laboratory of Osteo-Articular and Dental Tissue Engineering, Group STEP “Skeletal tissue Engineering and Physiopathology”, Nantes, France
| | - Sophie Sourice
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- INSERM, U791, Laboratory of Osteo-Articular and Dental Tissue Engineering, Group STEP “Skeletal tissue Engineering and Physiopathology”, Nantes, France
| | - Kevin Biteau
- INSERM UMR1087, CNRS UMR6291, l’institut du thorax, Nantes, France
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
| | - Philippe Hulin
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- Cellular and Tissular Imaging Core Facility of Nantes University (MicroPICell), Nantes, France
| | - Virginie Forest
- INSERM UMR1087, CNRS UMR6291, l’institut du thorax, Nantes, France
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
| | - Pierre Weiss
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- CHU de Nantes, Nantes, France
- INSERM, U791, Laboratory of Osteo-Articular and Dental Tissue Engineering, Group STEP “Skeletal tissue Engineering and Physiopathology”, Nantes, France
| | - Jérôme Guicheux
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- INSERM, U791, Laboratory of Osteo-Articular and Dental Tissue Engineering, Group STEP “Skeletal tissue Engineering and Physiopathology”, Nantes, France
| | - Patricia Lemarchand
- INSERM UMR1087, CNRS UMR6291, l’institut du thorax, Nantes, France
- Université de Nantes, Structure Fédérative de Recherche Santé F. Bonamy, Nantes, France
- CHU de Nantes, Nantes, France
- * E-mail:
| |
Collapse
|
45
|
Portron S, Merceron C, Maillard N, Weiss P, Grimandi G, Vinatier C, Guicheux J, Gauthier O. Cellules souches et biomatériaux injectables pour la médecine régénératrice du cartilage : le consortium « chondrograft ». Ing Rech Biomed 2012. [DOI: 10.1016/j.irbm.2012.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
46
|
Merceron C, Portron S, Masson M, Lesoeur J, Fellah BH, Gauthier O, Geffroy O, Weiss P, Guicheux J, Vinatier C. The Effect of Two- and Three-Dimensional Cell Culture on the Chondrogenic Potential of Human Adipose-Derived Mesenchymal Stem Cells after Subcutaneous Transplantation with an Injectable Hydrogel. Cell Transplant 2011; 20:1575-88. [DOI: 10.3727/096368910x557191] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage is an avascular tissue composed of chondrocytes, a unique cell type responsible for abundant matrix synthesis and maintenance. When damaged, it never heals spontaneously under physiological circumstances. Therefore, the delivery of mesenchymal stem cells using hydrogel has been considered for cartilage repair. This study aims at investigating the influence of in vitro chondrogenic differentiation of human adipose tissue-derived stem cells (hATSCs) on in vivo cartilage formation when associated with a cellulose-based self-setting hydrogel (Si-HPMC). hATSCs were characterized for their proliferation, surface marker expression, and multipotency. The in vitro chondrogenic potential of hATSCs cultured within Si-HPMC in control or chondrogenic medium was evaluated by measuring COL2A1, ACAN, SOX9, and COMP expression by real-time PCR. Alcian blue and type II collagen staining were also performed. To determine whether in vitro chondrogenically differentiated hATSCs may give rise to cartilage in vivo, cells differentiated as a monolayer or in pellets were finally associated with Si-HPMC and implanted subcutaneously into nude mice. Cartilage formation was assessed histologically by alcian blue and type II collagen staining. Our data demonstrate that hATSCs exhibited proliferation and self-renewal. hATSCs also expressed typical stem cell surface markers and were able to differentiate towards the adipogenic, osteogenic, and chondrogenic lineages. Real-time PCR and histological analysis indicated that Si-HPMC enabled chondrogenic differentiation of hATSCs in inductive medium, as demonstrated by increased expression of chondrogenic markers. In addition, histological analysis of implants showed that chondrogenically differentiated hATSCs (monolayers or pellets) have the ability to form cartilaginous tissue, as indicated by the presence of sulphated glycosaminoglycans and type II collagen. This study therefore suggests that an in vitro induction of hATSCs in 2D was sufficient to obtain cartilaginous tissue formation in vivo. Si-HPMC associated with autologous hATSCs could thus be a significant tool for regenerative medicine in the context of cartilage damage.
Collapse
Affiliation(s)
- Christophe Merceron
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
| | - Sophie Portron
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
| | - Martial Masson
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
| | - Julie Lesoeur
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
| | - Borhane Hakim Fellah
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
- Ecole Nationale vétérinaire de Nantes (ONIRIS), Service de chirurgie expérimentale (CRIP), Atlanpôle-La Chantrerie, Nantes Cedex 3, France
- Graftys SA-Eiffel Park-Bâtiment D-415, Aix en Provence Cedex 3, France
| | - Olivier Gauthier
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
- Ecole Nationale vétérinaire de Nantes (ONIRIS), Service de chirurgie expérimentale (CRIP), Atlanpôle-La Chantrerie, Nantes Cedex 3, France
| | - Olivier Geffroy
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
- Ecole Nationale vétérinaire de Nantes (ONIRIS), service de chirurgie équine, Atlanpôle-La Chantrerie, Nantes Cedex 3, France
| | - Pierre Weiss
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
| | - Jérôme Guicheux
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
| | - Claire Vinatier
- INSERM (Institut National de la Santé et de la Recherche Médicale), UMRS 791, Université de Nantes, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, Group STEP “Skeletal tissue Engineering and Physiopathology,” Faculté de chirurgie dentaire, Nantes Cedex 1, France
- PRES-UNAM, UFR Odontologie, Université de Nantes, Nantes, France
- Graftys SA-Eiffel Park-Bâtiment D-415, Aix en Provence Cedex 3, France
| |
Collapse
|
47
|
Albrecht C, Tichy B, Nürnberger S, Hosiner S, Zak L, Aldrian S, Marlovits S. Gene expression and cell differentiation in matrix-associated chondrocyte transplantation grafts: a comparative study. Osteoarthritis Cartilage 2011; 19:1219-27. [PMID: 21820068 DOI: 10.1016/j.joca.2011.07.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 06/29/2011] [Accepted: 07/05/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Although scaffold composition and architecture are considered to be important parameters for tissue engineering, their influence on gene expression and cell differentiation is rarely investigated in scaffolds used for matrix-associated autologous chondrocyte transplantation (MACT). In this study we have therefore comparatively analyzed the gene expression of important chondrogenic markers in four clinical applied cell-graft systems with very different scaffold characteristics. METHODS Residuals (n=165) of four different transplant types (MACI®, Hyalograft®C, CaReS® and Novocart®3D) were collected during surgery and analyzed for Col1, Col2, aggrecan, versican, melanoma inhibitory activity (MIA) and IL-1β by real-time PCR. Scaffold and cell morphology were evaluated by histology and electron microscopy. RESULTS Despite the cultivation on 3D scaffolds, the cell differentiation on all transplant types didn't reach the levels of native cartilage. Gene expression highly differed between the transplant types. The highest differentiation of cells (Col2/Col1 ratio) was found in CaReS®, followed by Novocart®3D, Hyalograft®C and MACI®. IL-1β expression also exhibited high differences between the scaffolds showing low expression levels in Novocart®3D and CaReS® and higher expression levels in MACI® and Hyalograft®C. CONCLUSIONS Our data indicate that scaffold characteristics as well as culture conditions highly influence gene expression in cartilage transplants and that these parameters may have profound impact on the tissue regeneration after MACT.
Collapse
Affiliation(s)
- C Albrecht
- Medical University of Vienna, Department of Traumatology, Center for Joint and Cartilage, Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Bone tissue engineering is a promising solution for patients with bone defects that require reconstruction. This regenerative therapy consists in culturing osteogenic cells on a biodegradable substrate to obtain a bio-hybrid construct that will stimulate bone healing after implantation. This multidisciplinary technology nevertheless requires further development before it can become routine clinical practice. One challenge is to achieve three-dimensional seeding and osteogenic commitment of mesenchymal stem cells on biomaterials under sterile and reproducible conditions. For this purpose, different dynamic culture systems have been developed. This paper reviews recent advances in the field of bioreactors for bone tissue engineering. The purpose of such systems is to improve nutrient delivery to the cells and generate shear stress that may promote cell differentiation into osteoblastic phenotypes. A brief overview of the value of computational fluid dynamics for understanding the cell environment is also provided. Finally, some proposals are made regarding the use of bioreactors as safe and controllable devices that will help commit cells and biomaterials for the regeneration of bone tissue.
Collapse
|
49
|
Ruan Y, Lin H, Yao J, Chen Z, Shao Z. Preparation of 3D fibroin/chitosan blend porous scaffold for tissue engineering via a simplified method. Macromol Biosci 2011; 11:419-26. [PMID: 21218404 DOI: 10.1002/mabi.201000392] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Indexed: 11/08/2022]
Abstract
In this work, we developed a simple and flexible method to manufacture a 3D porous scaffold based on the blend of regenerated silk fibroin (RSF) and chitosan (CS). No crosslinker or other toxic reagents were used in this method. The pores of resulted 3D scaffolds were connected with each other, and their sizes could be easily controlled by the concentration of the mixed solution. Compared with pure RSF scaffolds, the water absorptivities of these RSF/CS blend scaffolds with significantly enhanced mechanical properties were greatly increased. The results of MTT and RT-PCR tests indicated that the chondrocytes grew very well in these blend RSF/CS porous scaffolds. This suggested that the RSF/CS blend scaffold prepared by this new method could be a promising candidate for applications in tissue engineering.
Collapse
Affiliation(s)
- Yuhui Ruan
- The Key Laboratory of Molecular Engineering of Polymers of MOE, Department of Macromolecular Science, The Laboratory of Advanced Materials, Fudan University, Shanghai 200433, PR China
| | | | | | | | | |
Collapse
|
50
|
Sayed KE, Haisch A, John T, Marzahn U, Lohan A, Müller RD, Kohl B, Ertel W, Stoelzel K, Schulze-Tanzil G. Heterotopic Autologous Chondrocyte Transplantation—A Realistic Approach to Support Articular Cartilage Repair? TISSUE ENGINEERING PART B-REVIEWS 2010; 16:603-16. [DOI: 10.1089/ten.teb.2010.0167] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Karym El Sayed
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Andreas Haisch
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Thilo John
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Ulrike Marzahn
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Anke Lohan
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Riccarda D. Müller
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Benjamin Kohl
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Wolfgang Ertel
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Katharina Stoelzel
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin, Berlin, Germany
| | - Gundula Schulze-Tanzil
- Department of Trauma and Reconstructive Surgery, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|