1
|
Vincent R, Foston M, Hammond WB, Collins GL, Arinzeh TL. Synthesis and Characterization of Glycosaminoglycan Mimetic Variants to Promote Chondrogenesis. ACS OMEGA 2025; 10:4505-4515. [PMID: 39959109 PMCID: PMC11822690 DOI: 10.1021/acsomega.4c08084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/18/2024] [Accepted: 01/14/2025] [Indexed: 02/18/2025]
Abstract
Tissue engineering strategies to treat cartilage damage remain inadequate because of the difficulty in regenerating fully functional cartilage tissue. Sulfated glycosaminoglycans (GAGs), which are found in the native extracellular matrix, are known to interact with growth factors and, thus, promote chondrocyte function. Native GAGs have been explored as viable scaffold materials for tissue repair applications. However, it is unclear what structural features in GAGs are critical for promoting chondrogenesis. Therefore, this study generated GAG mimetics that vary in glycosidic linkage geometry and monomer ring substitution and were evaluated for their effect on mesenchymal stem cell (MSC) chondrogenesis and their potential use in cartilage tissue engineering applications. GAG mimetics were synthesized from cellulose (pSC), starch (SS), and chitin (ChS). pSC has beta-glycosidic linkages, SS has alpha-glycosidic linkages, and ChS has beta-glycosidic linkages and monomers that consist of the amide derivative of glucose. Evaluated in soluble form in MSC pellet cultures, pSC and SS enhanced MSC chondrogenic differentiation as measured by the deposition of chondrogenic matrix components, collagen type II and GAG normalized to the cell number, over ChS and the control culture media (without GAG mimetics). The higher degree of sulfation (DOS) in both the pSC and SS also had an effect on the relative collagen type II deposition and GAG production. These data suggest that beta- and alpha-glycosidic linkages are favorable for promoting chondrogenesis. This study demonstrates the potential of semisynthetic GAG mimetics for chondrogenic differentiation, where structural features should be considered for cartilage repair applications.
Collapse
Affiliation(s)
- Richard Vincent
- Department
of Biomedical Engineering, New Jersey Institute
of Technology, Newark, New Jersey 07102, United States
| | - Marcus Foston
- Department
of Energy, Environmental & Chemical Engineering, Washington University in Saint Louis, Saint Louis, Missouri 63130, United States
| | - Willis B. Hammond
- Department
of Biomedical Engineering, New Jersey Institute
of Technology, Newark, New Jersey 07102, United States
| | - George L. Collins
- Department
of Biomedical Engineering, New Jersey Institute
of Technology, Newark, New Jersey 07102, United States
| | | |
Collapse
|
2
|
Zigan C, Benito Alston C, Chatterjee A, Solorio L, Chan DD. Characterization of Composite Agarose-Collagen Hydrogels for Chondrocyte Culture. Ann Biomed Eng 2025; 53:120-132. [PMID: 39277549 PMCID: PMC11782374 DOI: 10.1007/s10439-024-03613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
To elucidate the mechanisms of cellular mechanotransduction, it is necessary to employ biomaterials that effectively merge biofunctionality with appropriate mechanical characteristics. Agarose and collagen separately are common biopolymers used in cartilage mechanobiology and mechanotransduction studies but lack features that make them ideal for functional engineered cartilage. In this study, agarose is blended with collagen type I to create hydrogels with final concentrations of 4% w/v or 2% w/v agarose with 2 mg/mL collagen. We hypothesized that the addition of collagen into a high-concentration agarose hydrogel does not diminish mechanical properties. Acellular and cell-laden studies were completed to assess rheologic and compressive properties, contraction, and structural homogeneity in addition to cell proliferation and sulfated glycosaminoglycan production. Over 21 days in culture, cellular 4% agarose-2 mg/mL collagen I hydrogels seeded with primary murine chondrocytes displayed structural and bulk mechanical behaviors that did not significantly alter from 4% agarose-only hydrogels, cell proliferation, and continual glycosaminoglycan production, indicating promise toward the development of an effective hydrogel for chondrocyte mechanotransduction and mechanobiology studies.
Collapse
Affiliation(s)
- Clarisse Zigan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | | | - Aritra Chatterjee
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Department of Mechanical Engineering, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Hyderabad, Telangana, India
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Deva D Chan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
3
|
Diab RG, Deeb G, Roda R, Karam M, Faraj M, Harajli M, Damiati LA, Mhanna R. Maintaining the Cartilage Phenotype of Late-Passage Chondrocytes Using Salidroside, TGF-β, and Sulfated Alginate for Cartilage Tissue Engineering Applications. Int J Mol Sci 2024; 25:13623. [PMID: 39769386 PMCID: PMC11727720 DOI: 10.3390/ijms252413623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/07/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
The limited self-repair capacity of cartilage due to its avascular and aneural nature leads to minimal regenerative ability. Autologous chondrocyte transplantation (ACT) is a popular treatment for cartilage defects but faces challenges due to chondrocyte dedifferentiation in later passages, which results in undesirable fibroblastic phenotypes. A promising treatment for cartilage injuries and diseases involves tissue engineering using cells (e.g., chondrocytes), scaffolds (e.g., Alginate Sulfate (AlgSulf)), and biochemical signals (e.g., Salidroside and TGF-β). This study focuses on investigating the effects of AlgSulf scaffolds with varying degrees of sulfation, Salidroside, and TGF-β on the proliferation, viability, and phenotype maintenance of chondrocytes. The findings demonstrate that AlgSulf films with a degree of sulfation (DS) = 2, treated with a combination of Salidroside and TGF-β, significantly enhanced chondrocyte proliferation (p < 0.001 and p < 0.0001 in P2 and P4, respectively), preserved round cell morphology, and maintained cartilage-specific gene expression (Col2, Aggrecans, and SOX9) while downregulating fibroblastic markers (Col1, MMP13, IL-1β, and IL-6). Our findings suggest the potential of this combination for enhancing cartilage regeneration in tissue engineering applications.
Collapse
Affiliation(s)
- Rita G. Diab
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - George Deeb
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Rena Roda
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Mia Karam
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Marwa Faraj
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Mohamad Harajli
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| |
Collapse
|
4
|
Chu YY, Hikita A, Asawa Y, Hoshi K. Advancements in chondrocyte 3-dimensional embedded culture: Implications for tissue engineering and regenerative medicine. Biomed J 2024; 48:100786. [PMID: 39236979 PMCID: PMC12018037 DOI: 10.1016/j.bj.2024.100786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Cartilage repair necessitates regenerative medicine because of the unreliable healing mechanism of cartilage. To yield a sufficient number of cells for transplantation, chondrocytes must be expanded in culture. However, in 2D culture, chondrocytes tend to lose their distinctive phenotypes and functionalities after serial passage, thereby limiting their efficacy for tissue engineering purposes. The mechanism of dedifferentiation in 2D culture can be attributed to various factors, including abnormal nuclear strength, stress-induced mitochondrial impairment, chromatin remodeling, ERK-1/2 and the p38/mitogen-activated protein kinase (MAPK) signaling pathway. These mechanisms collectively contribute to the loss of chondrocyte phenotype and reduced production of cartilage-specific extracellular matrix (ECM) components. Chondrocyte 3D culture methods have emerged as promising solutions to prevent dedifferentiation. Techniques, such as scaffold-based culture and scaffold-free approaches, provide chondrocytes with a more physiologically relevant environment, promoting their differentiation and matrix synthesis. These methods have been used in cartilage tissue engineering to create engineered cartilage constructs for transplantation and joint repair. However, chondrocyte 3D culture still has limitations, such as low viability and proliferation rate, and also difficulties in passage under 3D condition. These indicate challenges of obtaining a sufficient number of chondrocytes for large-scale tissue production. To address these issues, ongoing studies of many research groups have been focusing on refining culture conditions, optimizing scaffold materials, and exploring novel cell sources such as stem cells to enhance the quality and quantity of engineered cartilage tissues. Although obstacles remain, continuous endeavors to enhance culture techniques and overcome limitations offer a promising outlook for the advancement of more efficient strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Yu-Ying Chu
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Plastic and Reconstructive Surgery, Craniofacial Research Centre, Chang Gung Memorial Hospital at Linko, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
5
|
Alizadeh S, Ameri Z, Daemi H, Pezeshki-Modaress M. Sulfated polysaccharide as biomimetic biopolymers for tissue engineering scaffolds fabrication: Challenges and opportunities. Carbohydr Polym 2024; 336:122124. [PMID: 38670755 DOI: 10.1016/j.carbpol.2024.122124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/31/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024]
Abstract
Sulfated polysaccharides play important roles in tissue engineering applications because of their high growth factor preservation ability and their native-like biological features. There are different sulfated polysaccharides based on different repeating units in the carbohydrate backbone, the position of the sulfate group, and the sulfation degree of the polysaccharide. These led to various sulfated polymers with different negative charge densities and resultant structure-property relationships. Since numerous reports are presented related to sulfated polysaccharide applications in tissue engineering, it is crucial to review the role of effective physicochemical and biological parameters in their usage; as well as their structure-property relationships. Within this review, we focused on the effect of naturally occurring and synthetic sulfated polysaccharides in tissue engineering applications reported in the last years, highlighting the challenges of the scaffold fabrication process, the position, and the degree of sulfate on biomedical activity. Additionally, we discussed their use in numerous in vitro and in vivo model systems.
Collapse
Affiliation(s)
- Sanaz Alizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ameri
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamad Pezeshki-Modaress
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Plastic and Reconstructive surgery, Hazrat Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Zhao F, Qiu Y, Liu W, Zhang Y, Liu J, Bian L, Shao L. Biomimetic Hydrogels as the Inductive Endochondral Ossification Template for Promoting Bone Regeneration. Adv Healthc Mater 2024; 13:e2303532. [PMID: 38108565 DOI: 10.1002/adhm.202303532] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Repairing critical size bone defects (CSBD) is a major clinical challenge and requires effective intervention by biomaterial scaffolds. Inspired by the fact that the cartilaginous template-based endochondral ossification (ECO) process is crucial to bone healing and development, developing biomimetic biomaterials to promote ECO is recognized as a promising approach for repairing CSBD. With the unique highly hydrated 3D polymeric network, hydrogels can be designed to closely emulate the physiochemical properties of cartilage matrix to facilitate ECO. In this review, the various preparation methods of hydrogels possessing the specific physiochemical properties required for promoting ECO are introduced. The materiobiological impacts of the physicochemical properties of hydrogels, such as mechanical properties, topographical structures and chemical compositions on ECO, and the associated molecular mechanisms related to the BMP, Wnt, TGF-β, HIF-1α, FGF, and RhoA signaling pathways are further summarized. This review provides a detailed coverage on the materiobiological insights required for the design and preparation of hydrogel-based biomaterials to facilitate bone regeneration.
Collapse
Affiliation(s)
- Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yonghao Qiu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Wenjing Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Jia Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, P. R. China
| |
Collapse
|
7
|
Karam M, Faraj M, Jaffa MA, Jelwan J, Aldeen KS, Hassan N, Mhanna R, Jaffa AA. Development of alginate and alginate sulfate/polycaprolactone nanoparticles for growth factor delivery in wound healing therapy. Biomed Pharmacother 2024; 175:116750. [PMID: 38749174 DOI: 10.1016/j.biopha.2024.116750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024] Open
Abstract
Connective tissue growth factor (CTGF) holds great promise for enhancing the wound healing process; however, its clinical application is hindered by its low stability and the challenge of maintaining its effective concentration at the wound site. Herein, we developed novel double-emulsion alginate (Alg) and heparin-mimetic alginate sulfate (AlgSulf)/polycaprolactone (PCL) nanoparticles (NPs) for controlled CTGF delivery to promote accelerated wound healing. The NPs' physicochemical properties, cytocompatibility, and wound healing activity were assessed on immortalized human keratinocytes (HaCaT), primary human dermal fibroblasts (HDF), and a murine cutaneous wound model. The synthesized NPs had a minimum hydrodynamic size of 200.25 nm. Treatment of HaCaT and HDF cells with Alg and AlgSulf2.0/PCL NPs did not show any toxicity when used at concentrations <50 µg/mL for up to 72 h. Moreover, the NPs' size was not affected by elevated temperatures, acidic pH, or the presence of a protein-rich medium. The NPs have slow lysozyme-mediated degradation implying that they have an extended tissue retention time. Furthermore, we found that treatment of HaCaT and HDF cells with CTGF-loaded Alg and AlgSulf2.0/PCL NPs, respectively, induced rapid cell migration (76.12% and 79.49%, P<0.05). Finally, in vivo studies showed that CTGF-loaded Alg and AlgSulf2.0/PCL NPs result in the fastest and highest wound closure at the early and late stages of wound healing, respectively (36.49%, P<0.001 on day 1; 90.45%, P<0.05 on day 10), outperforming free CTGF. Double-emulsion NPs based on Alg or AlgSulf represent a viable strategy for delivering heparin-binding GF and other therapeutics, potentially aiding various disease treatments.
Collapse
Affiliation(s)
- Mia Karam
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut Lebanon
| | - Marwa Faraj
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut Lebanon
| | - Miran A Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, P.O. Box 11-0236, Beirut, Lebanon
| | - Joseph Jelwan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut Lebanon
| | - Kawthar Sharaf Aldeen
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut Lebanon
| | - Nadine Hassan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut Lebanon
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Beirut Lebanon.
| |
Collapse
|
8
|
Karaca MA, Kancagi DD, Ozbek U, Ovali E, Gok O. Betulin Stimulates Osteogenic Differentiation of Human Osteoblasts-Loaded Alginate-Gelatin Microbeads. Bioengineering (Basel) 2024; 11:553. [PMID: 38927789 PMCID: PMC11201098 DOI: 10.3390/bioengineering11060553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Osteoporosis, a terminal illness, has emerged as a global public health problem in recent years. The long-term use of bone anabolic drugs to treat osteoporosis causes multi-morbidity in elderly patients. Alternative therapies, such as allogenic and autogenic tissue grafts, face important issues, such as a limited source of allogenic grafts and tissue rejection in autogenic grafts. However, stem cell therapy has been shown to increase bone regeneration and decrease osteoporotic bone formation. Stem cell therapy combined with betulin (BET) supplementation might be adequate for bone remodeling and new bone tissue generation. In this study, the effect of BET on the viability and osteogenic differentiation of hFOB 1.19 cells was investigated. The cells were encapsulated in alginate-gelatin (AlGel) microbeads. In vitro tests were conducted during the 12 d of incubation. While BET showed cytotoxic activity (>1 µM) toward non-encapsulated hFOB 1.19 cells, encapsulated cells retained their functionality for up to 12 days, even at 5 µM BET. Moreover, the expression of osteogenic markers indicates an enhanced osteo-inductive effect of betulin on encapsulated hFOB 1.19, compared to the non-encapsulated cell culture. The 3D micro-environment of the AlGel microcapsules successfully protects the hFOB 1.19 cells against BET cytotoxicity, allowing BET to improve the mineralization and differentiation of osteoblast cells.
Collapse
Affiliation(s)
- Mehmet Ali Karaca
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Derya Dilek Kancagi
- Acibadem Labcell Cellular Therapy Laboratory, 34752 Istanbul, Turkey; (D.D.K.); (E.O.)
| | - Ugur Ozbek
- Medical Genetics Department, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Ercument Ovali
- Acibadem Labcell Cellular Therapy Laboratory, 34752 Istanbul, Turkey; (D.D.K.); (E.O.)
| | - Ozgul Gok
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
9
|
Bermudez-Lekerika P, Crump KB, Wuertz-Kozak K, Le Maitre CL, Gantenbein B. Sulfated Hydrogels as Primary Intervertebral Disc Cell Culture Systems. Gels 2024; 10:330. [PMID: 38786247 PMCID: PMC11121347 DOI: 10.3390/gels10050330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
The negatively charged extracellular matrix plays a vital role in intervertebral disc tissues, providing specific cues for cell maintenance and tissue hydration. Unfortunately, suitable biomimetics for intervertebral disc regeneration are lacking. Here, sulfated alginate was investigated as a 3D culture material due to its similarity to the charged matrix of the intervertebral disc. Precursor solutions of standard alginate, or alginate with 0.1% or 0.2% degrees of sulfation, were mixed with primary human nucleus pulposus cells, cast, and cultured for 14 days. A 0.2% degree of sulfation resulted in significantly decreased cell density and viability after 7 days of culture. Furthermore, a sulfation-dependent decrease in DNA content and metabolic activity was evident after 14 days. Interestingly, no significant differences in cell density and viability were observed between surface and core regions for sulfated alginate, unlike in standard alginate, where the cell number was significantly higher in the core than in the surface region. Due to low cell numbers, phenotypic evaluation was not achieved in sulfated alginate biomaterial. Overall, standard alginate supported human NP cell growth and viability superior to sulfated alginate; however, future research on phenotypic properties is required to decipher the biological properties of sulfated alginate in intervertebral disc cells.
Collapse
Affiliation(s)
- Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland; (P.B.-L.); (K.B.C.)
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Katherine B. Crump
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland; (P.B.-L.); (K.B.C.)
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA;
- Spine Center, Schön Klinik München Harlaching Academic Teaching Hospital, Spine Research Institute, Paracelsus Private Medical University Salzburg (Austria), 81547 Munich, Germany
| | - Christine L. Le Maitre
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK;
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland; (P.B.-L.); (K.B.C.)
- Inselspital, Department of Orthopedic Surgery & Traumatology, Medical Faculty, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
10
|
Surman F, Asadikorayem M, Weber P, Weber D, Zenobi-Wong M. Ionically annealed zwitterionic microgels for bioprinting of cartilaginous constructs. Biofabrication 2024; 16:025004. [PMID: 38176081 DOI: 10.1088/1758-5090/ad1b1f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/04/2024] [Indexed: 01/06/2024]
Abstract
Foreign body response (FBR) is a pervasive problem for biomaterials used in tissue engineering. Zwitterionic hydrogels have emerged as an effective solution to this problem, due to their ultra-low fouling properties, which enable them to effectively inhibit FBRin vivo. However, no versatile zwitterionic bioink that allows for high resolution extrusion bioprinting of tissue implants has thus far been reported. In this work, we introduce a simple, novel method for producing zwitterionic microgel bioink, using alginate methacrylate (AlgMA) as crosslinker and mechanical fragmentation as a microgel fabrication method. Photocrosslinked hydrogels made of zwitterionic carboxybetaine acrylamide (CBAA) and sulfobetaine methacrylate (SBMA) are mechanically fragmented through meshes with aperture diameters of 50 and 90µm to produce microgel bioink. The bioinks made with both microgel sizes showed excellent rheological properties and were used for high-resolution printing of objects with overhanging features without requiring a support structure or support bath. The AlgMA crosslinker has a dual role, allowing for both primary photocrosslinking of the bulk hydrogel as well as secondary ionic crosslinking of produced microgels, to quickly stabilize the printed construct in a calcium bath and to produce a microporous scaffold. Scaffolds showed ∼20% porosity, and they supported viability and chondrogenesis of encapsulated human primary chondrocytes. Finally, a meniscus model was bioprinted, to demonstrate the bioink's versatility at printing large, cell-laden constructs which are stable for furtherin vitroculture to promote cartilaginous tissue production. This easy and scalable strategy of producing zwitterionic microgel bioink for high resolution extrusion bioprinting allows for direct cell encapsulation in a microporous scaffold and has potential forin vivobiocompatibility due to the zwitterionic nature of the bioink.
Collapse
Affiliation(s)
- František Surman
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, 8032 Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
11
|
Mutch AL, Ferro V, A A, Grøndahl L. Synthesis of sulfated alginate from its tributylammonium salt: Comparing the sulfating agents H 2SO 4-DCC and SO 3·py. Carbohydr Polym 2024; 324:121488. [PMID: 37985083 DOI: 10.1016/j.carbpol.2023.121488] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 11/22/2023]
Abstract
Direct comparison of the sulfating agents H2SO4-DCC and SO3·py for the synthesis of sulfated alginate (S-Alg) as well as detailed characterisation of the products that form is lacking. This study involving three researchers used the tributylammonium salt of alginate (T-Alg) as a common substrate for the sulfation reactions. It was found that the use of H2SO4-DCC resulted in poor control of the degree of sulfation (DS) and that the S-Alg polymers contained nitrogen (determined by elemental analysis) as a result of formation of an unwanted N-acylurea adduct. Additionally, a large reduction in chain length was confirmed. In contrast, the use of SO3·py gave reasonable control over DS, resulted in high yields, showed no contamination and no clear change in chain length. Detailed characterisation of such S-Alg polymers by 1H NMR, 13C NMR and 1H,13C-HSQC NMR confirmed sulfation at C2 and C3 with a preference for C2.
Collapse
Affiliation(s)
- Alexandra L Mutch
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland 4072, Australia
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland 4072, Australia
| | - Anitha A
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland 4072, Australia.
| | - Lisbeth Grøndahl
- School of Chemistry and Molecular Biosciences, The University of Queensland, Cooper Road, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
12
|
Qi J, Wu H, Liu G. Novel Strategies for Spatiotemporal and Controlled BMP-2 Delivery in Bone Tissue Engineering. Cell Transplant 2024; 33:9636897241276733. [PMID: 39305020 PMCID: PMC11418245 DOI: 10.1177/09636897241276733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 09/25/2024] Open
Abstract
Bone morphogenetic protein-2 (BMP-2) has been commercially approved by the Food and Drug Administration for use in bone defects and diseases. BMP-2 promotes osteogenic differentiation of mesenchymal stem cells. In bone tissue engineering, BMP-2 incorporated into scaffolds can be used for stimulating bone regeneration in organoid construction, drug testing platforms, and bone transplants. However, the high dosage and uncontrollable release rate of BMP-2 challenge its clinical application, mainly due to the short circulation half-life of BMP-2, microbial contamination in bone extracellular matrix hydrogel, and the delivery method. Moreover, in clinical translation, the requirement of high doses of BMP-2 for efficacy poses challenges in cost and safety. Based on these, novel strategies should ensure that BMP-2 is delivered precisely to the desired location within the body, regulating the timing of BMP-2 release to coincide with the bone healing process, as well as release BMP-2 in a controlled manner to optimize its therapeutic effect and minimize side effects. This review highlights improvements in bone tissue engineering applying spatiotemporal and controlled BMP-2 delivery, including molecular engineering, biomaterial modification, and synergistic therapy, aiming to provide references for future research and clinical trials.
Collapse
Affiliation(s)
- Jingqi Qi
- Department of Orthopedics, Third Xiangya Hospital of Central South University, Changsha, China
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hongwei Wu
- Department of Orthopedics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Gengyan Liu
- Department of Orthopedics, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
13
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
14
|
Menezes R, Vincent R, Osorno L, Hu P, Arinzeh TL. Biomaterials and tissue engineering approaches using glycosaminoglycans for tissue repair: Lessons learned from the native extracellular matrix. Acta Biomater 2023; 163:210-227. [PMID: 36182056 PMCID: PMC10043054 DOI: 10.1016/j.actbio.2022.09.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 01/30/2023]
Abstract
Glycosaminoglycans (GAGs) are an important component of the extracellular matrix as they influence cell behavior and have been sought for tissue regeneration, biomaterials, and drug delivery applications. GAGs are known to interact with growth factors and other bioactive molecules and impact tissue mechanics. This review provides an overview of native GAGs, their structure, and properties, specifically their interaction with proteins, their effect on cell behavior, and their mechanical role in the ECM. GAGs' function in the extracellular environment is still being understood however, promising studies have led to the development of medical devices and therapies. Native GAGs, including hyaluronic acid, chondroitin sulfate, and heparin, have been widely explored in tissue engineering and biomaterial approaches for tissue repair or replacement. This review focuses on orthopaedic and wound healing applications. The use of GAGs in these applications have had significant advances leading to clinical use. Promising studies using GAG mimetics and future directions are also discussed. STATEMENT OF SIGNIFICANCE: Glycosaminoglycans (GAGs) are an important component of the native extracellular matrix and have shown promise in medical devices and therapies. This review emphasizes the structure and properties of native GAGs, their role in the ECM providing biochemical and mechanical cues that influence cell behavior, and their use in tissue regeneration and biomaterial approaches for orthopaedic and wound healing applications.
Collapse
Affiliation(s)
- Roseline Menezes
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Richard Vincent
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Laura Osorno
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Phillip Hu
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Treena Livingston Arinzeh
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States; Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States.
| |
Collapse
|
15
|
Baei P, Daemi H, Aramesh F, Baharvand H, Eslaminejad MB. Advances in mechanically robust and biomimetic polysaccharide-based constructs for cartilage tissue engineering. Carbohydr Polym 2023; 308:120650. [PMID: 36813342 DOI: 10.1016/j.carbpol.2023.120650] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
The purpose of cartilage tissue engineering is to provide artificial constructs with biological functions and mechanical features that resemble native tissue to improve tissue regeneration. Biochemical characteristics of the cartilage extracellular matrix (ECM) microenvironment provide a platform for researchers to develop biomimetic materials for optimal tissue repair. Due to the structural similarity of polysaccharides into physicochemical characteristics of cartilage ECM, these natural polymers capture special attention for developing biomimetic materials. The mechanical properties of constructs play a crucial influence in load-bearing cartilage tissues. Moreover, the addition of appropriate bioactive molecules to these constructs can promote chondrogenesis. Here, we discuss polysaccharide-based constructs that can be used to create substitutes for cartilage regeneration. We intend to focus on newly developed bioinspired materials, fine-tuning the mechanical properties of constructs, the design of carriers loaded by chondroinductive agents, and development of appropriate bioinks as a bioprinting approach for cartilage regeneration.
Collapse
Affiliation(s)
- Payam Baei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran.
| | - Fatemeh Aramesh
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University ofTehran, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
16
|
Lin YY, Kuan CY, Chang CT, Chuang MH, Syu WS, Zhang KL, Lee CH, Lin PC, Dong GC, Lin FH. 3D-Cultured Adipose-Derived Stem Cell Spheres Using Calcium-Alginate Scaffolds for Osteoarthritis Treatment in a Mono-Iodoacetate-Induced Rat Model. Int J Mol Sci 2023; 24:ijms24087062. [PMID: 37108239 PMCID: PMC10138691 DOI: 10.3390/ijms24087062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease that causes pain, cartilage deformation, and joint inflammation. Mesenchymal stem cells (MSCs) are potential therapeutic agents for OA treatment. However, the 2D culture of MSCs could potentially affect their characteristics and functionality. In this study, calcium-alginate (Ca-Ag) scaffolds were prepared for human adipose-derived stem cell (hADSC) proliferation with a homemade functionally closed process bioreactor system; the feasibility of cultured hADSC spheres in heterologous stem cell therapy for OA treatment was then evaluated. hADSC spheres were collected from Ca-Ag scaffolds by removing calcium ions via ethylenediaminetetraacetic acid (EDTA) chelation. In this study, 2D-cultured individual hADSCs or hADSC spheres were evaluated for treatment efficacy in a monosodium iodoacetate (MIA)-induced OA rat model. The results of gait analysis and histological sectioning showed that hADSC spheres were more effective at relieving arthritis degeneration. The results of serological and blood element analyses of hADSC-treated rats indicated that the hADSC spheres were a safe treatment in vivo. This study demonstrates that hADSC spheres are a promising treatment for OA and can be applied to other stem cell therapies or regenerative medical treatments.
Collapse
Affiliation(s)
- Yu-Ying Lin
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Che-Yung Kuan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 10087, Taiwan
| | - Chia-Tien Chang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Ming-Hsi Chuang
- College of Management, Chung Hwa University, Hsinchu 30012, Taiwan
| | - Wan-Sin Syu
- Gwo Xi Stem Cell Applied Technology, Hsinchu 30261, Taiwan
| | - Kai-Ling Zhang
- Gwo Xi Stem Cell Applied Technology, Hsinchu 30261, Taiwan
- College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Chia-Hsin Lee
- Gwo Xi Stem Cell Applied Technology, Hsinchu 30261, Taiwan
| | - Po-Cheng Lin
- Gwo Xi Stem Cell Applied Technology, Hsinchu 30261, Taiwan
| | - Guo-Chung Dong
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Feng-Huei Lin
- Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County 35053, Taiwan
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 10087, Taiwan
| |
Collapse
|
17
|
Zhang W, Kuss M, Yan Y, Shi W. Dynamic Alginate Hydrogel as an Antioxidative Bioink for Bioprinting. Gels 2023; 9:gels9040312. [PMID: 37102924 PMCID: PMC10137987 DOI: 10.3390/gels9040312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
3D bioprinting holds great potential for use in tissue engineering to treat degenerative joint disorders, such as osteoarthritis. However, there is a lack of multifunctional bioinks that can not only support cell growth and differentiation, but also offer protection to cells against injuries caused by the elevated oxidative stress; this conditions is a common characteristic of the microenvironment of the osteoarthritis disease. To mitigate oxidative stress-induced cellular phenotype change and malfunction, an anti-oxidative bioink derived from an alginate dynamic hydrogel was developed in this study. The alginate dynamic hydrogel gelated quickly via the dynamic covalent bond between the phenylboronic acid modified alginate (Alg-PBA) and poly (vinyl alcohol) (PVA). It presented good self-healing and shear-thinning abilities because of the dynamic feature. The dynamic hydrogel supported long-term growth of mouse fibroblasts after stabilization with a secondary ionic crosslinking between introduced calcium ions and the carboxylate group in the alginate backbone. In addition, the dynamic hydrogel showed good printability, resulting in the fabrication of scaffolds with cylindrical and grid structures with good structural fidelity. Encapsulated mouse chondrocytes maintained high viability for at least 7 days in the bioprinted hydrogel after ionic crosslinking. Most importantly, in vitro studies implied that the bioprinted scaffold could reduce the intracellular oxidative stress for embedded chondrocytes under H2O2 exposure; it could also protect the chondrocytes from H2O2-induced downregulation of extracellular matrix (ECM) relevant anabolic genes (ACAN and COL2) and upregulation of a catabolic gene (MMP13). In summary, the results suggest that the dynamic alginate hydrogel can be applied as a versatile bioink for the fabrication of 3D bioprinted scaffolds with an innate antioxidative ability; this technique is expected to improve the regenerative efficacy of cartilage tissues for the treatment of joint disorders.
Collapse
Affiliation(s)
- Wenhai Zhang
- Orthopedic Department, Tianjin Hospital, Tianjin 300211, China
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Yan
- Healthcare Security Office, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
18
|
Mousavi SS, Keshvari H, Daemi H. Partial sulfation of gellan gum produces cytocompatible, body temperature-responsive hydrogels. Int J Biol Macromol 2023; 235:123525. [PMID: 36841392 DOI: 10.1016/j.ijbiomac.2023.123525] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/27/2023]
Abstract
Gellan gum (GG) is a biodegradable polysaccharide and forms thermosensitive hydrogels by a helix-mediated mechanism. Unfortunately, the wide use of GG in tissue engineering has been restricted due to its dramatically higher gelation temperature than normal body temperature. Here, we show that partial sulfation of GG affords a cytocompatible body temperature-responsive hydrogel with an interesting thermoreversibility at 42 °C. The partial sulfation of GG was confirmed by FTIR, EDX and elemental analyses. The sulfated GGs (SGGs) had a higher swelling ratio and degradation in PBS compared to the neat GG. Based on the results of rheometry analysis, the SGG with a degree of sulfation of 0.27 (H3 sample) showed a gelation temperature close to the physiological temperature. In addition, the drop in mechanical properties of SGGs was compensated by a further calcium-mediated ionic crosslinking, where Young's modulus of H3 increased from 10.6 ± 1.9 kPa up to 38.4 ± 5.5 kPa. Finally, we showed that the partial sulfation reaction of GG is a simple and mild strategy to modify chemical structure of GG, and to produce a cytocompatible, body temperature-responsive hydrogel compared to other modifying reactions such as oxidation reaction.
Collapse
Affiliation(s)
- Seyed Saeed Mousavi
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran; Department of Cell Engineering, Stem Cells and Developmental Biology, Cell Science Research Center, ACECR, Royan institute, Tehran, Iran
| | - Hamid Keshvari
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Hamed Daemi
- Department of Cell Engineering, Stem Cells and Developmental Biology, Cell Science Research Center, ACECR, Royan institute, Tehran, Iran.
| |
Collapse
|
19
|
Exploration of Dual Ionic Cross-Linked Alginate Hydrogels Via Cations of Varying Valences towards Wound Healing. Polymers (Basel) 2022; 14:polym14235192. [PMID: 36501587 PMCID: PMC9738749 DOI: 10.3390/polym14235192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
This study explored the synergistic effects of simultaneously using calcium and gallium cations in the cross-linking of alginate, detailing its effects on the characteristics of alginate compared to its single cation counterparts. The primary goal is to determine if there are any synergistic effects associated with the utilisation of multiple multivalent cations in polymer cross-linking and whether or not it could therefore be used in pharmaceutical applications such as wound healing. Given the fact divalent and trivalent cations have never been utilised together for cross-linking, an explanation for the mode of binding that occurs between the alginate and the cations during the cross-linking process and how it may affect the future applications of the polymer has been investigated. The calcium gallium alginate polymers were able to retain the antibacterial effects of gallium within the confines of the polymer matrix, possessing superior rheological properties, 6 times that of pure calcium and pure gallium, coupled with an improved swelling capacity that is 4 times higher than that of gallium alginate.
Collapse
|
20
|
Dye BR, Decker JT, Hein RF, Miller AJ, Huang S, Spence JR, Shea LD. Human Lung Organoid Culture in Alginate With and Without Matrigel to Model Development and Disease. Tissue Eng Part A 2022; 28:893-906. [PMID: 36029210 PMCID: PMC9805886 DOI: 10.1089/ten.tea.2022.0054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/12/2022] [Indexed: 01/13/2023] Open
Abstract
Human lung organoids (HLOs) are enabling the study of human lung development and disease by modeling native organ tissue structure, cellular composition, and cellular organization. In this report, we demonstrate that HLOs derived from human pluripotent stem cells cultured in alginate, a fully defined nonanimal product substrate, exhibit enhanced cellular differentiation compared with HLOs cultured in the commercially available Matrigel. More specifically, we observed an earlier onset and increase in the number of multiciliated cells, along with mucus producing MUC5AC+ goblet-like cells that were not observed in HLOs cultured in Matrigel. The epithelium in alginate-grown HLOs was organized in a pseudostratified epithelium with airway basal cells lining the basal lamina, but with the apical surface of cells on the exterior of the organoid. We further observed that HLOs cultured in Matrigel exhibited mesenchymal overgrowth that was not present in alginate cultures. The containment of the mesenchyme within HLOs in alginate enabled modeling of key features of idiopathic pulmonary fibrosis (IPF) by treatment with transforming growth factor β (TGFβ). TGFβ treatment resulted in morphological changes including an increase in mesenchymal growth, increased expression of IPF markers, and decreased numbers of alveolar-like cells. This culture system provides a model to study the interaction of the mesenchyme with the epithelium during lung development and diseased states such as IPF.
Collapse
Affiliation(s)
- Briana R. Dye
- Department of Biomedical Engineering, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph T. Decker
- Department of Biomedical Engineering, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Renee F.C. Hein
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alyssa J. Miller
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sha Huang
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R. Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
García-Briega MI, Ródenas-Rochina J, Martins LA, Lanceros-Méndez S, Gallego Ferrer G, Sempere A, Gómez Ribelles JL. Stability of Biomimetically Functionalised Alginate Microspheres as 3D Support in Cell Cultures. Polymers (Basel) 2022; 14:4282. [PMID: 36297867 PMCID: PMC9611185 DOI: 10.3390/polym14204282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022] Open
Abstract
Alginate hydrogels can be used to develop a three-dimensional environment in which various cell types can be grown. Cross-linking the alginate chains using reversible ionic bonds opens up great possibilities for the encapsulation and subsequent release of cells or drugs. However, alginate also has a drawback in that its structure is not very stable in a culture medium with cellular activity. This work explored the stability of alginate microspheres functionalised by grafting specific biomolecules onto their surface to form microgels in which biomimetic microspheres surrounded the cells in the culture, reproducing the natural microenvironment. A study was made of the stability of the microgel in different typical culture media and the formation of polyelectrolyte multilayers containing polylysine and heparin. Multiple myeloma cell proliferation in the culture was tested in a bioreactor under gentle agitation.
Collapse
Affiliation(s)
- María Inmaculada García-Briega
- Centre for Biomaterials and Tissue Engineering (CBIT) Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain
| | - Joaquín Ródenas-Rochina
- Centre for Biomaterials and Tissue Engineering (CBIT) Universitat Politècnica de València, 46022 Valencia, Spain
| | - Luis Amaro Martins
- Centre for Biomaterials and Tissue Engineering (CBIT) Universitat Politècnica de València, 46022 Valencia, Spain
| | - Senentxu Lanceros-Méndez
- Centre of Physics, Universidade Do Minho, 4710-057 Braga, Portugal
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Gloria Gallego Ferrer
- Centre for Biomaterials and Tissue Engineering (CBIT) Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain
| | - Amparo Sempere
- Grupo de Investigación en Hematología, Instituto de Investigación Sanitaria La Fe (IISLAFE), 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, 28029 Madrid, Spain
| | - José Luís Gómez Ribelles
- Centre for Biomaterials and Tissue Engineering (CBIT) Universitat Politècnica de València, 46022 Valencia, Spain
- Biomedical Research Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 46022 Valencia, Spain
| |
Collapse
|
22
|
Miri L, Irani S, Pezeshki-Modaress M, Daemi H, Atyabi SM. Guiding mesenchymal stem cells differentiation into chondrocytes using sulfated alginate/cold atmospheric plasma modified polycaprolactone nanofibrous scaffold. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04476-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Bertsch P, Diba M, Mooney DJ, Leeuwenburgh SCG. Self-Healing Injectable Hydrogels for Tissue Regeneration. Chem Rev 2022; 123:834-873. [PMID: 35930422 PMCID: PMC9881015 DOI: 10.1021/acs.chemrev.2c00179] [Citation(s) in RCA: 279] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biomaterials with the ability to self-heal and recover their structural integrity offer many advantages for applications in biomedicine. The past decade has witnessed the rapid emergence of a new class of self-healing biomaterials commonly termed injectable, or printable in the context of 3D printing. These self-healing injectable biomaterials, mostly hydrogels and other soft condensed matter based on reversible chemistry, are able to temporarily fluidize under shear stress and subsequently recover their original mechanical properties. Self-healing injectable hydrogels offer distinct advantages compared to traditional biomaterials. Most notably, they can be administered in a locally targeted and minimally invasive manner through a narrow syringe without the need for invasive surgery. Their moldability allows for a patient-specific intervention and shows great prospects for personalized medicine. Injected hydrogels can facilitate tissue regeneration in multiple ways owing to their viscoelastic and diffusive nature, ranging from simple mechanical support, spatiotemporally controlled delivery of cells or therapeutics, to local recruitment and modulation of host cells to promote tissue regeneration. Consequently, self-healing injectable hydrogels have been at the forefront of many cutting-edge tissue regeneration strategies. This study provides a critical review of the current state of self-healing injectable hydrogels for tissue regeneration. As key challenges toward further maturation of this exciting research field, we identify (i) the trade-off between the self-healing and injectability of hydrogels vs their physical stability, (ii) the lack of consensus on rheological characterization and quantitative benchmarks for self-healing injectable hydrogels, particularly regarding the capillary flow in syringes, and (iii) practical limitations regarding translation toward therapeutically effective formulations for regeneration of specific tissues. Hence, here we (i) review chemical and physical design strategies for self-healing injectable hydrogels, (ii) provide a practical guide for their rheological analysis, and (iii) showcase their applicability for regeneration of various tissues and 3D printing of complex tissues and organoids.
Collapse
Affiliation(s)
- Pascal Bertsch
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands
| | - Mani Diba
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - David J. Mooney
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - Sander C. G. Leeuwenburgh
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,
| |
Collapse
|
24
|
Gradišnik L, Maver U, Gole B, Bunc G, Voršič M, Ravnik J, Šmigoc T, Bošnjak R, Velnar T. The Endplate Role in Degenerative Disc Disease Research: The Isolation of Human Chondrocytes from Vertebral Endplate-An Optimised Protocol. Bioengineering (Basel) 2022; 9:137. [PMID: 35447697 PMCID: PMC9029037 DOI: 10.3390/bioengineering9040137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Degenerative disc disease is a progressive and chronic disorder with many open questions regarding its pathomorphological mechanisms. In related studies, in vitro organ culture systems are becoming increasingly essential as a replacement option for laboratory animals. Live disc cells are highly appealing to study the possible mechanisms of intervertebral disc (IVD) degeneration. To study the degenerative processes of the endplate chondrocytes in vitro, we established a relatively quick and easy protocol for isolating human chondrocytes from the vertebral endplates. METHODS The fragments of human lumbar endplates following lumbar fusion were collected, cut, ground and partially digested with collagenase I in Advanced DMEM/F12 with 5% foetal bovine serum. The sediment was harvested, and cells were seeded in suspension, supplemented with special media containing high nutrient levels. Morphology was determined with phalloidin staining and the characterisation for collagen I, collagen II and aggrecan with immunostaining. RESULTS The isolated cells retained viability in appropriate laboratory conditions and proliferated quickly. The confluent culture was obtained after 14 days. Six to 8 h after seeding, attachments were observed, and proliferation of the isolated cells followed after 12 h. The cartilaginous endplate chondrocytes were stable with a viability of up to 95%. Pheno- and geno-typic analysis showed chondrocyte-specific expression, which decreased with passages. CONCLUSIONS The reported cell isolation process is simple, economical and quick, allowing establishment of a viable long-term cell culture. The availability of a vertebral endplate cell model will permit the study of cell properties, biochemical aspects, the potential of therapeutic candidates for the treatment of disc degeneration, and toxicology studies in a well-controlled environment.
Collapse
Affiliation(s)
- Lidija Gradišnik
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (L.G.); (U.M.)
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (L.G.); (U.M.)
| | - Boris Gole
- Centre for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia;
| | - Gorazd Bunc
- Department of Neurosurgery, University Medical Centre Maribor, 2000 Maribor, Slovenia; (G.B.); (M.V.); (J.R.); (T.Š.)
| | - Matjaž Voršič
- Department of Neurosurgery, University Medical Centre Maribor, 2000 Maribor, Slovenia; (G.B.); (M.V.); (J.R.); (T.Š.)
| | - Janez Ravnik
- Department of Neurosurgery, University Medical Centre Maribor, 2000 Maribor, Slovenia; (G.B.); (M.V.); (J.R.); (T.Š.)
| | - Tomaž Šmigoc
- Department of Neurosurgery, University Medical Centre Maribor, 2000 Maribor, Slovenia; (G.B.); (M.V.); (J.R.); (T.Š.)
| | - Roman Bošnjak
- Department of Neurosurgery, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| | - Tomaž Velnar
- Department of Neurosurgery, University Medical Centre Maribor, 2000 Maribor, Slovenia; (G.B.); (M.V.); (J.R.); (T.Š.)
- Department of Neurosurgery, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia;
| |
Collapse
|
25
|
Philippe V, Laurent A, Hirt-Burri N, Abdel-Sayed P, Scaletta C, Schneebeli V, Michetti M, Brunet JF, Applegate LA, Martin R. Retrospective Analysis of Autologous Chondrocyte-Based Cytotherapy Production for Clinical Use: GMP Process-Based Manufacturing Optimization in a Swiss University Hospital. Cells 2022; 11:1016. [PMID: 35326468 PMCID: PMC8947208 DOI: 10.3390/cells11061016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Cultured autologous human articular chondrocyte (HAC) implantation has been extensively investigated for safe and effective promotion of structural and functional restoration of knee cartilage lesions. HAC-based cytotherapeutic products for clinical use must be manufactured under an appropriate quality assurance system and follow good manufacturing practices (GMP). A prospective clinical trial is ongoing in the Lausanne University Hospital, where the HAC manufacturing processes have been implemented internally. Following laboratory development and in-house GMP transposition of HAC cell therapy manufacturing, a total of 47 patients have been treated to date. The main aim of the present study was to retrospectively analyze the available manufacturing records of the produced HAC-based cytotherapeutic products, outlining the inter-individual variability existing among the 47 patients regarding standardized transplant product preparation. These data were used to ameliorate and to ensure the continued high quality of cytotherapeutic care in view of further clinical investigations, based on the synthetic analyses of existing GMP records. Therefore, a renewed risk analysis-based process definition was performed, with specific focus set on process parameters, controls, targets, and acceptance criteria. Overall, high importance of the interdisciplinary collaboration and of the manufacturing process robustness was underlined, considering the high variability (i.e., quantitative, functional) existing between the treated patients and between the derived primary HAC cell types.
Collapse
Affiliation(s)
- Virginie Philippe
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (V.S.); (R.M.)
| | - Alexis Laurent
- Manufacturing Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland;
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
- DLL Bioengineering, Discovery Learning Program, STI School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Valentine Schneebeli
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (V.S.); (R.M.)
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Murielle Michetti
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
| | - Jean-François Brunet
- Cell Production Center, Service of Pharmacy, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland;
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (N.H.-B.); (P.A.-S.); (C.S.); (M.M.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Robin Martin
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland; (V.S.); (R.M.)
| |
Collapse
|
26
|
Zare P, Pezeshki-Modaress M, Davachi SM, Chahsetareh H, Simorgh S, Asgari N, Haramshahi MA, Alizadeh R, Bagher Z, Farhadi M. An additive manufacturing-based 3D printed poly ɛ-caprolactone/alginate sulfate/extracellular matrix construct for nasal cartilage regeneration. J Biomed Mater Res A 2022; 110:1199-1209. [PMID: 35098649 DOI: 10.1002/jbm.a.37363] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/28/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
Various composite scaffolds with different fabrication techniques have been applied in cartilage tissue engineering. In this study, poly ɛ-caprolactone (PCL) was printed by fused deposition modeling method, and the prepared scaffold was filled with Alginate (Alg): Alginate-Sulfate (Alg-Sul) hydrogel to provide a better biomimetic environment and emulate the structure of glycosaminoglycans properly. Furthermore, to enhance chondrogenesis, different concentrations of decellularized extracellular matrix (dECM) were added to the hydrogel. For cellular analyses, the adipose-derived mesenchymal stem cells were seeded on the hydrogel and the results of MTT assay, live/dead staining, and SEM images revealed that the scaffold with 1% dECM had better viscosity, cell viability, and proliferation. The study was conducted on the optimized scaffold (1% dECM) to determine mechanical characteristics, chondrogenic differentiation, and results demonstrated that the scaffold showed mechanical similarity to the native nasal cartilage tissue along with possessing appropriate biochemical features, which makes this new formulation based on PCL/dECM/Alg:Alg-Sul a promising candidate for further in-vivo studies.
Collapse
Affiliation(s)
- Pariya Zare
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | | | - Seyed Mohammad Davachi
- Department of Biology and Chemistry, Texas A&M International University, Laredo, Texas, USA
| | - Hadi Chahsetareh
- Department of Life Science Engineering, Faculty of New Science and Technologies, University of Tehran, Tehran, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negin Asgari
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Amin Haramshahi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rafieh Alizadeh
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohamad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
27
|
Klimek K, Tarczynska M, Truszkiewicz W, Gaweda K, Douglas TEL, Ginalska G. Freeze-Dried Curdlan/Whey Protein Isolate-Based Biomaterial as Promising Scaffold for Matrix-Associated Autologous Chondrocyte Transplantation-A Pilot In-Vitro Study. Cells 2022; 11:282. [PMID: 35053397 PMCID: PMC8773726 DOI: 10.3390/cells11020282] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 01/18/2023] Open
Abstract
The purpose of this pilot study was to establish whether a novel freeze-dried curdlan/whey protein isolate-based biomaterial may be taken into consideration as a potential scaffold for matrix-associated autologous chondrocyte transplantation. For this reason, this biomaterial was initially characterized by the visualization of its micro- and macrostructures as well as evaluation of its mechanical stability, and its ability to undergo enzymatic degradation in vitro. Subsequently, the cytocompatibility of the biomaterial towards human chondrocytes (isolated from an orthopaedic patient) was assessed. It was demonstrated that the novel freeze-dried curdlan/whey protein isolate-based biomaterial possessed a porous structure and a Young's modulus close to those of the superficial and middle zones of cartilage. It also exhibited controllable degradability in collagenase II solution over nine weeks. Most importantly, this biomaterial supported the viability and proliferation of human chondrocytes, which maintained their characteristic phenotype. Moreover, quantitative reverse transcription PCR analysis and confocal microscope observations revealed that the biomaterial may protect chondrocytes from dedifferentiation towards fibroblast-like cells during 12-day culture. Thus, in conclusion, this pilot study demonstrated that novel freeze-dried curdlan/whey protein isolate-based biomaterial may be considered as a potential scaffold for matrix-associated autologous chondrocyte transplantation.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (W.T.); (G.G.)
| | - Marta Tarczynska
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
| | - Wieslaw Truszkiewicz
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (W.T.); (G.G.)
| | - Krzysztof Gaweda
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
| | - Timothy E. L. Douglas
- Engineering Department, Lancaster University, Gillow Avenue, Lancaster LA 1 4YW, UK;
- Materials Science Institute (MSI), Lancaster University, Lancaster LA 1 4YW, UK
| | - Grazyna Ginalska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (W.T.); (G.G.)
| |
Collapse
|
28
|
Lazarus E, Bermudez-Lekerika P, Farchione D, Schofield T, Howard S, Mambetkadyrov I, Lamoca M, Rivero IV, Gantenbein B, Lewis CL, Wuertz-Kozak K. Sulfated Hydrogels in Intervertebral Disc and Cartilage Research. Cells 2021; 10:cells10123568. [PMID: 34944076 PMCID: PMC8700363 DOI: 10.3390/cells10123568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 01/07/2023] Open
Abstract
Hydrogels are commonly used for the 3D culture of musculoskeletal cells. Sulfated hydrogels, which have seen a growing interest over the past years, provide a microenvironment that help maintain the phenotype of chondrocytes and chondrocyte-like cells and can be used for sustained delivery of growth factors and other drugs. Sulfated hydrogels are hence valuable tools to improve cartilage and intervertebral disc tissue engineering. To further advance the utilization of these hydrogels, we identify and summarize the current knowledge about different sulfated hydrogels, highlight their beneficial effects in cartilage and disc research, and review the biofabrication processes most suitable to secure best quality assurance through deposition fidelity, repeatability, and attainment of biocompatible morphologies.
Collapse
Affiliation(s)
- Emily Lazarus
- Department of Industrial and Systems Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (E.L.); (I.V.R.)
| | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
| | - Daniel Farchione
- Inamori School of Engineering, Alfred University, Alfred, NY 14802, USA;
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Taylor Schofield
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Sloan Howard
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Iskender Mambetkadyrov
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Mikkael Lamoca
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Iris V. Rivero
- Department of Industrial and Systems Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (E.L.); (I.V.R.)
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, CH-3010 Bern, Switzerland
| | - Christopher L. Lewis
- Department of Manufacturing and Mechanical Engineering Technology, Rochester Institute of Technology, Rochester, NY 14632, USA;
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14632, USA; (T.S.); (S.H.); (I.M.); (M.L.)
- Schoen Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (AU), 81547 Munich, Germany
- Correspondence: ; Tel.: +1-585-475-7355
| |
Collapse
|
29
|
Alginate Modification and Lectin-Conjugation Approach to Synthesize the Mucoadhesive Matrix. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112411818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alginates are natural anionic polyelectrolytes investigated in various biomedical applications, such as drug delivery, tissue engineering, and 3D bioprinting. Functionalization of alginates is one possible way to provide a broad range of requirements for those applications. A range of techniques, including esterification, amidation, acetylation, phosphorylation, sulfation, graft copolymerization, and oxidation and reduction, have been implemented for this purpose. The rationale behind these investigations is often the combination of such modified alginates with different molecules. Particularly promising are lectin conjugate macromolecules for lectin-mediated drug delivery, which enhance the bioavailability of active ingredients on a specific site. Most interesting for such application are alginate derivatives, because these macromolecules are more resistant to acidic and enzymatic degradation. This review will report recent progress in alginate modification and conjugation, focusing on alginate-lectin conjugation, which is proposed as a matrix for mucoadhesive drug delivery and provides a new perspective for future studies with these conjugation methods.
Collapse
|
30
|
Wang B, Chariyev-Prinz F, Burdis R, Eichholz K, Kelly DJ. Additive manufacturing of cartilage-mimetic scaffolds as off-the-shelf implants for joint regeneration. Biofabrication 2021; 14. [PMID: 34883477 DOI: 10.1088/1758-5090/ac41a0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/09/2021] [Indexed: 11/11/2022]
Abstract
Biomimetic scaffolds that provide a tissue-specific environment to cells are particularly promising for tissue engineering and regenerative medicine applications. The goal of this study was to integrate emerging additive manufacturing and biomaterial design strategies to produce articular cartilage (AC) mimetic scaffolds that could be used as 'off-the-shelf' implants for joint regeneration. To this end alginate sulfate, a sulfated glycosaminoglycan (sGAG) mimic, was used to functionalize porous alginate-based scaffolds and to support the sustained release of transforming growth factor-β3 (TGF-β3). Covalent crosslinking dramatically improved the elasticity of the alginate/alginate sulfate scaffolds, while scaffold architecture could be tailored using a directional freezing technique. Introducing such an anisotropic architecture was found to promote mesenchymal stem cell (MSC) infiltration into the scaffold and to direct the orientation of the deposited extracellular matrix, leading to the development of cartilage tissue with a biomimetic zonal architecture. In vitro experiments also demonstrated the capacity of the sulfated scaffolds to both enhance chondrogenesis of MSCs and to control the release of TGF-β3, leading to the development of a tissue rich in sGAG and type II collagen. The scaffolds were further reinforced with a 3D printed PLCL framework, leading to composite implants that were more elastic than those reinforced with PCL, and which better mimicked the bulk mechanical properties of native cartilage tissue. The ability of this composite scaffold to support chondrogenesis was then confirmed within a dynamic culture system. Altogether, these findings demonstrate the potential of such biomimetic scaffolds as putative 'single-stage' or 'off-the-shelf' strategies for articular cartilage regeneration.
Collapse
Affiliation(s)
- Bin Wang
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, IRELAND
| | - Farhad Chariyev-Prinz
- Trinity Biomedical Institute, Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Dublin, D02 PN40, IRELAND
| | - Ross Burdis
- Trinity Biomedical Institute, Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Dublin, D02 PN40, IRELAND
| | - Kian Eichholz
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, IRELAND
| | - Daniel John Kelly
- Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Parsons Building, Dublin 2, Dublin, IRELAND
| |
Collapse
|
31
|
Rosiak P, Latanska I, Paul P, Sujka W, Kolesinska B. Modification of Alginates to Modulate Their Physic-Chemical Properties and Obtain Biomaterials with Different Functional Properties. Molecules 2021; 26:7264. [PMID: 34885846 PMCID: PMC8659150 DOI: 10.3390/molecules26237264] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
Modified alginates have a wide range of applications, including in the manufacture of dressings and scaffolds used for regenerative medicine, in systems for selective drug delivery, and as hydrogel materials. This literature review discusses the methods used to modify alginates and obtain materials with new or improved functional properties. It discusses the diverse biological and functional activity of alginates. It presents methods of modification that utilize both natural and synthetic peptides, and describes their influence on the biological properties of the alginates. The success of functionalization depends on the reaction conditions being sufficient to guarantee the desired transformations and provide modified alginates with new desirable properties, but mild enough to prevent degradation of the alginates. This review is a literature description of efficient methods of alginate functionalization using biologically active ligands. Particular attention was paid to methods of alginate functionalization with peptides, because the combination of the properties of alginates and peptides leads to the obtaining of conjugates with properties resulting from both components as well as a completely new, different functionality.
Collapse
Affiliation(s)
- Piotr Rosiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Ilona Latanska
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Paulina Paul
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Witold Sujka
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| |
Collapse
|
32
|
Sulfated alginate/polycaprolactone double-emulsion nanoparticles for enhanced delivery of heparin-binding growth factors in wound healing applications. Colloids Surf B Biointerfaces 2021; 208:112105. [PMID: 34536674 DOI: 10.1016/j.colsurfb.2021.112105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/27/2021] [Accepted: 09/04/2021] [Indexed: 11/22/2022]
Abstract
Diabetic foot ulcers (DFUs) that are not effectively treated could lead to partial or complete lower limb amputations. The lack of connective tissue growth factor (CTGF) and insulin-like growth factor (IGF-I) in DFUs results in limited matrix deposition and poor tissue repair. To enhance growth factor (GF) availability in DFUs, heparin (HN)-mimetic alginate sulfate/polycaprolactone (AlgSulf/PCL) double emulsion nanoparticles (NPs) with high affinity and sustained release of CTGF and IGF-I were synthesized. The NPs size, encapsulation efficiency (EE), cytotoxicity, cellular uptake and wound healing capacity in immortalized primary human adult epidermal cells (HaCaT) were assessed. The sonication time and amplitude used for NPs synthesis enabled the production of particles with a minimum of 236 ± 25 nm diameter. Treatment of HaCaT cells with up to 50 μg mL-1 of NPs showed no cytotoxic effects after 72 h. The highest bovine serum albumin EE (94.6 %, P = 0.028) and lowest burst release were attained with AlgSulf/PCL. Moreover, cells treated with AlgSulf/CTGF (250 ng mL-1) exhibited the most rapid wound closure compared to controls while maintaining fibronectin synthesis. Double-emulsion NPs based on HN-mimetic AlgSulf represent a novel approach which can significantly enhance diabetic wound healing and can be expanded for applications requiring the delivery of other HN-binding GFs.
Collapse
|
33
|
Three Polymers from the Sea: Unique Structures, Directional Modifications, and Medical Applications. Polymers (Basel) 2021; 13:polym13152482. [PMID: 34372087 PMCID: PMC8348450 DOI: 10.3390/polym13152482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/24/2021] [Accepted: 07/06/2021] [Indexed: 01/07/2023] Open
Abstract
With the increase of wounds and body damage, the clinical demand for antibacterial, hemostatic, and repairable biomaterials is increasing. Various types of biomedical materials have become research hotspots. Of these, and among materials derived from marine organisms, the research and application of alginate, chitosan, and collagen are the most common. Chitosan is mainly used as a hemostatic material in clinical applications, but due to problems such as the poor mechanical strength of a single component, the general antibacterial ability, and fast degradation speed research into the extraction process and modification mainly focuses on the improvement of the above-mentioned ability. Similarly, the research and modification of sodium alginate, used as a material for hemostasis and the repair of wounds, is mainly focused on the improvement of cell adhesion, hydrophilicity, degradation speed, mechanical properties, etc.; therefore, there are fewer marine biological collagen products. The research mainly focuses on immunogenicity removal and mechanical performance improvement. This article summarizes the source, molecular structure, and characteristics of alginate, chitosan, and collagen from marine organisms; and introduces the biological safety, clinical efficacy, and mechanism of action of these materials, as well as their extraction processes and material properties. Their modification and other issues are also discussed, and their potential clinical applications are examined.
Collapse
|
34
|
Ahmad Raus R, Wan Nawawi WMF, Nasaruddin RR. Alginate and alginate composites for biomedical applications. Asian J Pharm Sci 2021; 16:280-306. [PMID: 34276819 PMCID: PMC8261255 DOI: 10.1016/j.ajps.2020.10.001] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/26/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Alginate is an edible heteropolysaccharide that abundantly available in the brown seaweed and the capsule of bacteria such as Azotobacter sp. and Pseudomonas sp. Owing to alginate gel forming capability, it is widely used in food, textile and paper industries; and to a lesser extent in biomedical applications as biomaterial to promote wound healing and tissue regeneration. This is evident from the rising use of alginate-based dressing for heavily exuding wound and their mass availability in the market nowadays. However, alginate also has limitation. When in contact with physiological environment, alginate could gelate into softer structure, consequently limits its potential in the soft tissue regeneration and becomes inappropriate for the usage related to load bearing body parts. To cater this problem, wide range of materials have been added to alginate structure, producing sturdy composite materials. For instance, the incorporation of adhesive peptide and natural polymer or synthetic polymer to alginate moieties creates an improved composite material, which not only possesses better mechanical properties compared to native alginate, but also grants additional healing capability and promote better tissue regeneration. In addition, drug release kinetic and cell viability can be further improved when alginate composite is used as encapsulating agent. In this review, preparation of alginate and alginate composite in various forms (fibre, bead, hydrogel, and 3D-printed matrices) used for biomedical application is described first, followed by the discussion of latest trend related to alginate composite utilization in wound dressing, drug delivery, and tissue engineering applications.
Collapse
Affiliation(s)
- Raha Ahmad Raus
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| | - Wan Mohd Fazli Wan Nawawi
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
- Nanoscience and Nanotechnology Research Group (NanoRG), International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| | - Ricca Rahman Nasaruddin
- Department of Biotechnology Engineering, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
- Nanoscience and Nanotechnology Research Group (NanoRG), International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia
| |
Collapse
|
35
|
Zare P, Pezeshki-Modaress M, Davachi SM, Zare P, Yazdian F, Simorgh S, Ghanbari H, Rashedi H, Bagher Z. Alginate sulfate-based hydrogel/nanofiber composite scaffold with controlled Kartogenin delivery for tissue engineering. Carbohydr Polym 2021; 266:118123. [PMID: 34044939 DOI: 10.1016/j.carbpol.2021.118123] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/15/2021] [Accepted: 04/25/2021] [Indexed: 11/27/2022]
Abstract
In this study, we fabricated two different arrangements of laminated composite scaffolds based on Alginate:Alginate sulfate hydrogel, PCL:Gelatin electrospun mat, and Kartogenin-PLGA nanoparticles (KGN-NPs). The optimized composite scaffold revealed a range of advantages such as improved mechanical features as well as less potential of damage (less dissipated energy), interconnected pores of hydrogel and fiber with adequate pore size, excellent swelling ratio, and controlled biodegradability. Furthermore, the synthesized KGN-NPs with spherical morphology were incorporated into the composite scaffold and exhibited a linear and sustained release of KGN within 30 days with desirable initial burst reduction (12% vs. 20%). Additionally, the cytotoxicity impact of the composite was evaluated. Resazurin assay and Live/Dead staining revealed that the optimized composite scaffold has no cytotoxic effect and could improve cell growth. Overall, according to the enhanced mechanical features, suitable environment for cellular growth, and sustained drug release, the optimized scaffold would be a good candidate for tissue regeneration.
Collapse
Affiliation(s)
- Pariya Zare
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | | | - Seyed Mohammad Davachi
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA.
| | - Pouria Zare
- Department of Civil & Environmental Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Fatemeh Yazdian
- Department of Life Science Engineering, Faculty of New Science and Technology, University of Tehran, Iran.
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hadi Ghanbari
- ENT and Head & Neck Research Center and Department, Hazrat Rasoul Akram Hospital, The Five Senses Health Institute, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Hamid Rashedi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Zohreh Bagher
- ENT and Head & Neck Research Center and Department, Hazrat Rasoul Akram Hospital, The Five Senses Health Institute, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
36
|
Xie A, Peng Y, Yao Z, Lu L, Ni T. Effect of a subset of adipose-derived stem cells isolated with liposome magnetic beads to promote cartilage repair. J Cell Mol Med 2021; 25:4204-4215. [PMID: 33768729 PMCID: PMC8093962 DOI: 10.1111/jcmm.16470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 03/05/2021] [Indexed: 12/20/2022] Open
Abstract
This study aimed to investigate the ability of CD146+ subset of ADSCs to repair cartilage defects. In this study, we prepared CD146+ liposome magnetic beads (CD146+LMB) to isolate CD146+ADSCs. The cells were induced for chondrogenic differentiation and verified by cartilage‐specific mRNA and protein expression. Then a mouse model of cartilage defect was constructed and treated by filling the induced cartilage cells into the damaged joint, to evaluate the function of such cells in the cartilage microenvironment. Our results demonstrated that the CD146+LMBs we prepared were uniform, small and highly stable, and cell experiments showed that the CD146+LMB has low cytotoxicity to the ADSCs. ADSCs isolated with CD146+LMB were all CD146+, CD105+, CD166+ and CD73+. After chondrogenic induction, the cells showed significantly increased expression of cartilage markers Sox9, collagen Ⅱ and aggrecan at protein level and significantly increased Sox9, collagen Ⅱ and aggrecan at mRNA level, and the protein expression and mRNA expression of CD146+ADSCs group were higher than those of ADSCs group. The CD146+ADSCs group showed superior tissue repair ability than the ADSCs group and blank control group in the animal experiment, as judged by gross observation, histological observation and histological scoring. The above results proved that CD146+LMB can successfully isolate the CD146+ADSCs, and after chondrogenic induction, these cells successfully promoted repair of articular cartilage defects, which may be a new direction of tissue engineering.
Collapse
Affiliation(s)
- Aiguo Xie
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinbo Peng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zuochao Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Habli Z, Deen NNA, Malaeb W, Mahfouz N, Mermerian A, Talhouk R, Mhanna R. Biomimetic sulfated glycosaminoglycans maintain differentiation markers of breast epithelial cells and preferentially inhibit proliferation of cancer cells. Acta Biomater 2021; 122:186-198. [PMID: 33444795 DOI: 10.1016/j.actbio.2020.12.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Glycosaminoglycans (GAG) are key elements involved in various physiological and pathological processes including cancer. Several GAG-based drugs have been developed showing significant results and potential use as cancer therapeutics. We previously reported that alginate sulfate (AlgSulf), a GAG-mimetic, reduces the proliferation of lung adenocarcinoma cells. In this study, we evaluated the preferential effect of AlgSulf on tumorigenic and nontumorigenic mammary epithelial cells in 2D, 3D, and coculture conditions. AlgSulf were synthesized with different degrees of sulfation (DSs) varying from 0 to 2.7 and used at 100 µg/mL on HMT-3522 S1 (S1) nontumorigenic mammary epithelial cells and their tumorigenic counterparts HMT-3522 T4-2 (T4-2) cells. The anti-tumor properties of AlgSulf were assessed using trypan blue and bromodeoxyuridine proliferation (BrdU) assays, immunofluorescence staining and transwell invasion assay. Binding of insulin and epidermal growth factor (EGF) to sulfated substrates was measured using QCM-D and ELISA. In 2D, the cell growth rate of cells treated with AlgSulf was consistently lower compared to untreated controls (p<0.001) and surpassed the effect of the native GAG heparin (positive control). In 3D, AlgSulf preferentially hindered the growth rate and the invasion potential of tumorigenic T4-2 nodules while maintaining the formation of differentiated polarized nontumorigenic S1 acini. The preferential growth inhibition of tumorigenic cells by AlgSulf was confirmed in a coculture system (p<0.001). In the ELISA assay, a trend of EGF binding was detected for sulfated polysaccharides while QCM-D analysis showed negligible binding of insulin and EGF to sulfated substrates. The preferential effect mediated by the mimetic sulfated GAGs on cancer cells may in part be growth factor dependent. Our findings suggest a potential anticancer therapeutic role of AlgSulf for the development of anticancer drugs.
Collapse
|
38
|
Ling Y, Zhang W, Wang P, Xie W, Yang W, Wang DA, Fan C. Three-dimensional (3D) hydrogel serves as a platform to identify potential markers of chondrocyte dedifferentiation by combining RNA sequencing. Bioact Mater 2021; 6:2914-2926. [PMID: 33718672 PMCID: PMC7917462 DOI: 10.1016/j.bioactmat.2021.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Dedifferentiation of chondrocyte greatly restricts its function and application, however, it is poorly understood except a small number of canonical markers. The non-cell-adhesive property endows polysaccharide hydrogel with the ability to maintain chondrocyte phenotype, which can serve as a platform to identify new molecular markers and therapeutic targets of chondrocyte dedifferentiation. In this study, the high-throughput RNA sequencing (RNA-seq) was first performed on articular chondrocytes at primary (P0) and passage 1 (P1) stages to explore the global alteration of gene expression along with chondrocyte dedifferentiation. Significantly, several potential marker genes, such as PFKFB3, KDM6B, had been identified via comparatively analyzing their expression in P0 and P1 chondrocytes as well as in 3D constructs (i.e. chondrocyte-laden alginate hydrogel and HA-MA hydrogel) at both mRNA and protein level. Besides, the changes in cellular morphology and enriched pathway of differentially expressed genes during chondrocyte dedifferentiation was studied in detail. This study developed the use of hydrogel as a platform to investigate chondrocyte dedifferentiation; the results provided new molecular markers and potential therapeutic targets of chondrocyte dedifferentiation.
Collapse
Affiliation(s)
- Yang Ling
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, PR China.,Department of Human Anatomy Histology and Embryology, School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Weiyuan Zhang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Peiyan Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Wanhua Xie
- The Precise Medicine Center, Shenyang Medical College, Shenyang, 110034, Liaoning, PR China
| | - Wei Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, PR China.,School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China.,Shenzhen Research Institute, City University of Hong Kong, Shenzhen Hi-tech Industrial Park, Shenzhen, Guangdong, 518057, PR China.,Karolinska Institute Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong, China
| | - Changjiang Fan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, Shandong, PR China.,Department of Human Anatomy Histology and Embryology, School of Basic Medicine, College of Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| |
Collapse
|
39
|
Laurent A, Abdel-Sayed P, Ducrot A, Hirt-Burri N, Scaletta C, Jaccoud S, Nuss K, de Buys Roessingh AS, Raffoul W, Pioletti D, von Rechenberg B, Applegate LA, Darwiche S. Development of Standardized Fetal Progenitor Cell Therapy for Cartilage Regenerative Medicine: Industrial Transposition and Preliminary Safety in Xenogeneic Transplantation. Biomolecules 2021; 11:250. [PMID: 33572428 PMCID: PMC7916236 DOI: 10.3390/biom11020250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/04/2021] [Accepted: 02/04/2021] [Indexed: 12/27/2022] Open
Abstract
Diverse cell therapy approaches constitute prime developmental prospects for managing acute or degenerative cartilaginous tissue affections, synergistically complementing specific surgical solutions. Bone marrow stimulation (i.e., microfracture) remains a standard technique for cartilage repair promotion, despite incurring the adverse generation of fibrocartilagenous scar tissue, while matrix-induced autologous chondrocyte implantation (MACI) and alternative autologous cell-based approaches may partly circumvent this effect. Autologous chondrocytes remain standard cell sources, yet arrays of alternative therapeutic biologicals present great potential for regenerative medicine. Cultured human epiphyseal chondro-progenitors (hECP) were proposed as sustainable, safe, and stable candidates for chaperoning cartilage repair or regeneration. This study describes the development and industrial transposition of hECP multi-tiered cell banking following a single organ donation, as well as preliminary preclinical hECP safety. Optimized cell banking workflows were proposed, potentially generating millions of safe and sustainable therapeutic products. Furthermore, clinical hECP doses were characterized as non-toxic in a standardized chorioallantoic membrane model. Lastly, a MACI-like protocol, including hECPs, was applied in a three-month GLP pilot safety evaluation in a caprine model of full-thickness articular cartilage defect. The safety of hECP transplantation was highlighted in xenogeneic settings, along with confirmed needs for optimal cell delivery vehicles and implantation techniques favoring effective cartilage repair or regeneration.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Épalinges, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Aurélie Ducrot
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
| | - Sandra Jaccoud
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, CH-2002 Neuchâtel, Switzerland;
| | - Katja Nuss
- Musculoskeletal Research Unit, Zurich Tierspital, University of Zurich, CH-8952 Schlieren, Switzerland; (K.N.); (B.v.R.)
| | - Anthony S. de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Dominique Pioletti
- Laboratory of Biomechanical Orthopedics, Ecole Polytechnique Fédérale de Lausanne, CH-2002 Neuchâtel, Switzerland;
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit, Zurich Tierspital, University of Zurich, CH-8952 Schlieren, Switzerland; (K.N.); (B.v.R.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1015 Lausanne, Switzerland; (A.L.); (P.A.-S.); (A.D.); (N.H.-B.); (C.S.); (S.J.); (L.A.A.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, Jiangsu, China
| | - Salim Darwiche
- Musculoskeletal Research Unit, Zurich Tierspital, University of Zurich, CH-8952 Schlieren, Switzerland; (K.N.); (B.v.R.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
40
|
Chen Z, Yu P, Miao Z, Zhang H, Xiao H, Xie J, Ding C, Li J. Sulfated alginate based complex for sustained calcitonin delivery and enhanced osteogenesis. Biomed Mater 2020; 16. [PMID: 33291091 DOI: 10.1088/1748-605x/abd1b9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
Direct medications of salmon calcitonin (sCT) through subcutaneous or intramuscular injection are limited for its low effeciency. Drug delivery systems with sustained delivery property and high bioactivity are imminently needed. In consideration of the clinic application, a cost-effective and effective carrier is demanded, which is still a challenge until now. In this study, a simple alginate/ alginate sulfate-sCT (Alg/AlgS-sCT) complex was succesfully constructed for sustained release of sCT. The negtively charged sulphate groups facilitate the bonding with sCT, which avoids the burst release of sCT and extends the release time up to 15 days (only 2 days for pure sCT). More importantly, the bioactivity of the released sCT is not affected during such long release time, suggesting a conformation similar to native sCT. In vitro analysis implies the biocompatibility of the complex. Moreover, the combination of AlgS and sCT synergistically impoved the osteogenic ability of MC3T3 cells, showing higher ALP level, intracellular and extracellular calcium ions concentrations. Note that the concentration of intracellular calcium ions displays 5.26 fold increments of control group after 10 days of incubation. We envision this simple yet effective system has potential applications in clinical trails and give inspiration for the design of other protein delivery system.
Collapse
Affiliation(s)
- Zhuoxin Chen
- Sichuan University, College of Polymer Science & Engineering No. 24 South Section 1, Yihuan Road, Chengdu, Sichuan, 610065, CHINA
| | - Peng Yu
- Sichuan University, College of Polymer Science & Engineering No. 24 South Section 1, Yihuan Road, Chengdu, 610065, CHINA
| | - Zhangshu Miao
- Sichuan University, College of Polymer Science & Engineering No. 24 South Section 1, Yihuan Road, Chengdu, Sichuan, 610065, CHINA
| | - Haochen Zhang
- Sichuan University, College of Polymer Science & Engineering No. 24 South Section 1, Yihuan Road, Chengdu, Sichuan, 610065, CHINA
| | - Hong Xiao
- Sichuan University, Department of Pain Management, West China Hospital, Sichuan University, No. 37, GuoXue Xiang, Chengdu, Sichuan, 610041, CHINA
| | - Jing Xie
- Sichuan University, College of Polymer Science & Engineering No. 24 South Section 1, Yihuan Road, Chengdu, Sichuan, 610065, CHINA
| | - Chunmei Ding
- College of Polymer Science & Engineering, Sichuan University, No. 24 South Section 1, Yihuan Road, Chengdu, China, Chengdu, 610065, CHINA
| | - Jianshu Li
- Sichuan University, College of Polymer Science & Engineering No. 24 South Section 1, Yihuan Road, Chengdu, 610065, CHINA
| |
Collapse
|
41
|
Laurent A, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh AS, Raffoul W, Applegate LA. Holistic Approach of Swiss Fetal Progenitor Cell Banking: Optimizing Safe and Sustainable Substrates for Regenerative Medicine and Biotechnology. Front Bioeng Biotechnol 2020; 8:557758. [PMID: 33195124 PMCID: PMC7644790 DOI: 10.3389/fbioe.2020.557758] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Safety, quality, and regulatory-driven iterative optimization of therapeutic cell source selection has constituted the core developmental bedrock for primary fetal progenitor cell (FPC) therapy in Switzerland throughout three decades. Customized Fetal Transplantation Programs were pragmatically devised as straightforward workflows for tissue procurement, traceability maximization, safety, consistency, and robustness of cultured progeny cellular materials. Whole-cell bioprocessing standardization has provided plethoric insights into the adequate conjugation of modern biotechnological advances with current restraining legislative, ethical, and regulatory frameworks. Pioneer translational advances in cutaneous and musculoskeletal regenerative medicine continuously demonstrate the therapeutic potential of FPCs. Extensive technical and clinical hindsight was gathered by managing pediatric burns and geriatric ulcers in Switzerland. Concomitant industrial transposition of dermal FPC banking, following good manufacturing practices, demonstrated the extensive potential of their therapeutic value. Furthermore, in extenso, exponential revalorization of Swiss FPC technology may be achieved via the renewal of integrative model frameworks. Consideration of both longitudinal and transversal aspects of simultaneous fetal tissue differential processing allows for a better understanding of the quasi-infinite expansion potential within multi-tiered primary FPC banking. Multiple fetal tissues (e.g., skin, cartilage, tendon, muscle, bone, lung) may be simultaneously harvested and processed for adherent cell cultures, establishing a unique model for sustainable therapeutic cellular material supply chains. Here, we integrated fundamental, preclinical, clinical, and industrial developments embodying the scientific advances supported by Swiss FPC banking and we focused on advances made to date for FPCs that may be derived from a single organ donation. A renewed model of single organ donation bioprocessing is proposed, achieving sustained standards and potential production of billions of affordable and efficient therapeutic doses. Thereby, the aim is to validate the core therapeutic value proposition, to increase awareness and use of standardized protocols for translational regenerative medicine, potentially impacting millions of patients suffering from cutaneous and musculoskeletal diseases. Alternative applications of FPC banking include biopharmaceutical therapeutic product manufacturing, thereby indirectly and synergistically enhancing the power of modern therapeutic armamentariums. It is hypothesized that a single qualifying fetal organ donation is sufficient to sustain decades of scientific, medical, and industrial developments, as technological optimization and standardization enable high efficiency.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Tec-Pharma SA, Bercher, Switzerland
- LAM Biotechnologies SA, Épalinges, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Anthony S. de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Wassim Raffoul
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Oxford Suzhou Center for Advanced Research, Science and Technology Co., Ltd., Oxford University, Suzhou, China
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
42
|
Irani S, Tavakkoli S, Pezeshki‐Modaress M, Taghavifar E, Mohammadali M, Daemi H. Electrospun nanofibrous alginate sulfate scaffolds promote mesenchymal stem cells differentiation to chondrocytes. J Appl Polym Sci 2020. [DOI: 10.1002/app.49868] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Shiva Irani
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Sajjad Tavakkoli
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | | | - Elham Taghavifar
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Marjan Mohammadali
- Department of Biology, Science and Research Branch Islamic Azad University Tehran Iran
| | - Hamed Daemi
- Department of Cell Engineering Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR Tehran Iran
| |
Collapse
|
43
|
Volkov AV, Muraev AA, Zharkova II, Voinova VV, Akoulina EA, Zhuikov VA, Khaydapova DD, Chesnokova DV, Menshikh KA, Dudun AA, Makhina TK, Bonartseva GA, Asfarov TF, Stamboliev IA, Gazhva YV, Ryabova VM, Zlatev LH, Ivanov SY, Shaitan KV, Bonartsev AP. Poly(3-hydroxybutyrate)/hydroxyapatite/alginate scaffolds seeded with mesenchymal stem cells enhance the regeneration of critical-sized bone defect. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:110991. [PMID: 32994018 DOI: 10.1016/j.msec.2020.110991] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/08/2020] [Accepted: 04/18/2020] [Indexed: 01/13/2023]
Abstract
A critical-sized calvarial defect in rats is employed to reveal the osteoinductive properties of biomaterials. In this study, we investigate the osteogenic efficiency of hybrid scaffolds based on composites of a biodegradable and biocompatible polymer, poly(3-hydroxybutyrate) (PHB) with hydroxyapatite (HA) filled with alginate (ALG) hydrogel containing mesenchymal stem cells (MSCs) on the regeneration of the critical-sized radial defect of the parietal bone in rats. The scaffolds based on PHB and PHB/HA with desired shapes were prepared by two-stage salt leaching technique using a mold obtained by three-dimensional printing. To obtain PHB/HA/ALG/MSC scaffolds seeded with MSCs, the scaffolds were filled with ALG hydrogel containing MSCs; acellular PHB/ALG and PHB/ALG filled with empty ALG hydrogel were prepared for comparison. The produced scaffolds have high porosity and irregular interconnected pore structure. PHB/HA scaffolds supported MSC growth and induced cell osteogenic differentiation in a regular medium in vitro that was manifested by an increase in ALP activity and expression of the CD45 phenotype marker. The data of computed tomography and histological studies showed 94% and 92%, respectively, regeneration of critical-sized calvarial bone defect in vivo at 28th day after implantation of MSC-seeded PHB/HA/ALG/MSC scaffolds with 3.6 times higher formation of the main amount of bone tissue at 22-28 days in comparison with acellular PHB/HA/ALG scaffolds that was shown at the first time by fluorescent microscopy using the original technique of intraperitoneal administration of fluorescent dyes to living postoperative rats. The obtained in vivo results can be associated with the MSC-friendly microstructure and in vitro osteogenic properties of PHB/HA base-scaffolds. Thus, the obtained data demonstrate the potential of MSCs encapsulated in the bioactive biopolymer/mineral/hydrogel scaffold to improve the bone regeneration process in critical-sized bone defects.
Collapse
Affiliation(s)
- Alexey V Volkov
- The Peoples' Friendship University of Russia, Miklukho-Maklaya St. 6, 117198 Moscow, Russia; N.N. Priorov National Medical Research Center of Traumatology and Orthopedics of the Ministry of Health of the Russian Federation, Priorova Str. 10, 127299 Moscow, Russia
| | - Alexander A Muraev
- The Peoples' Friendship University of Russia, Miklukho-Maklaya St. 6, 117198 Moscow, Russia; I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St. 8/2, 119991, Moscow, Russia
| | - Irina I Zharkova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia
| | - Vera V Voinova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia; A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia
| | - Elizaveta A Akoulina
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia; A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia
| | - Vsevolod A Zhuikov
- A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia
| | - Dolgor D Khaydapova
- Faculty of Soil Science, M.V.Lomonosov Moscow State University, Leninskie gory, 1, bld. 12, 119234 Moscow, Russia
| | - Dariana V Chesnokova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia
| | - Ksenia A Menshikh
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia
| | - Andrej A Dudun
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia; A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia
| | - Tatiana K Makhina
- A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia
| | - Garina A Bonartseva
- A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia
| | - Teymur F Asfarov
- The Peoples' Friendship University of Russia, Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| | - Ivan A Stamboliev
- The Peoples' Friendship University of Russia, Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| | - Yulia V Gazhva
- Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia
| | - Valentina M Ryabova
- Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, Minin and Pozharsky Sq. 10/1, 603005 Nizhny Novgorod, Russia
| | - Lubomir H Zlatev
- The Peoples' Friendship University of Russia, Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| | - Sergey Y Ivanov
- The Peoples' Friendship University of Russia, Miklukho-Maklaya St. 6, 117198 Moscow, Russia; I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St. 8/2, 119991, Moscow, Russia
| | - Konstantin V Shaitan
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia
| | - Anton P Bonartsev
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bld. 12, 119234 Moscow, Russia; A.N.Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld. 2, 119071 Moscow, Russia.
| |
Collapse
|
44
|
Moxon SR, Ferreira MJ, dos Santos P, Popa B, Gloria A, Katsarava R, Tugushi D, Serra AC, Hooper NM, Kimber SJ, Fonseca AC, Domingos MAN. A Preliminary Evaluation of the Pro-Chondrogenic Potential of 3D-Bioprinted Poly(ester Urea) Scaffolds. Polymers (Basel) 2020; 12:E1478. [PMID: 32630145 PMCID: PMC7408263 DOI: 10.3390/polym12071478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Degeneration of articular cartilage (AC) is a common healthcare issue that can result in significantly impaired function and mobility for affected patients. The avascular nature of the tissue strongly burdens its regenerative capacity contributing to the development of more serious conditions such as osteoarthritis. Recent advances in bioprinting have prompted the development of alternative tissue engineering therapies for the generation of AC. Particular interest has been dedicated to scaffold-based strategies where 3D substrates are used to guide cellular function and tissue ingrowth. Despite its extensive use in bioprinting, the application of polycaprolactone (PCL) in AC is, however, restricted by properties that inhibit pro-chondrogenic cell phenotypes. This study proposes the use of a new bioprintable poly(ester urea) (PEU) material as an alternative to PCL for the generation of an in vitro model of early chondrogenesis. The polymer was successfully printed into 3D constructs displaying adequate substrate stiffness and increased hydrophilicity compared to PCL. Human chondrocytes cultured on the scaffolds exhibited higher cell viability and improved chondrogenic phenotype with upregulation of genes associated with type II collagen and aggrecan synthesis. Bioprinted PEU scaffolds could, therefore, provide a potential platform for the fabrication of bespoke, pro-chondrogenic tissue engineering constructs.
Collapse
Affiliation(s)
- Samuel R. Moxon
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK; (S.R.M.); (N.M.H.)
| | - Miguel J.S. Ferreira
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK; (M.J.S.F.); (B.P.)
| | - Patricia dos Santos
- Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal; (P.d.S.); (A.C.S.)
| | - Bogdan Popa
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK; (M.J.S.F.); (B.P.)
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials—National Research Council of Italy, V.le J.F. Kennedy 54—Mostra d’Oltremare Pad. 20, 80125 Naples, Italy;
| | - Ramaz Katsarava
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, 240, David Aghmashenebeli Alley, Tbilisi 0159, Georgia; (R.K.); (D.T.)
| | - David Tugushi
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, 240, David Aghmashenebeli Alley, Tbilisi 0159, Georgia; (R.K.); (D.T.)
| | - Armenio C. Serra
- Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal; (P.d.S.); (A.C.S.)
| | - Nigel M. Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK; (S.R.M.); (N.M.H.)
| | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK;
| | - Ana C. Fonseca
- Centre for Mechanical Engineering, Materials and Processes, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal; (P.d.S.); (A.C.S.)
| | - Marco A. N. Domingos
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK;
- The Henry Royce Institute, The University of Manchester, Alan Turing Building, Oxford Road, Manchester M13 9PL, UK
| |
Collapse
|
45
|
Distler T, McDonald K, Heid S, Karakaya E, Detsch R, Boccaccini AR. Ionically and Enzymatically Dual Cross-Linked Oxidized Alginate Gelatin Hydrogels with Tunable Stiffness and Degradation Behavior for Tissue Engineering. ACS Biomater Sci Eng 2020; 6:3899-3914. [PMID: 33463325 DOI: 10.1021/acsbiomaterials.0c00677] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hydrogels that allow for the successful long-term in vitro culture of cell-biomaterial systems to enable the maturation of tissue engineering constructs are highly relevant in regenerative medicine. Naturally derived polysaccharide-based hydrogels promise to be one material group with enough versatility and chemical functionalization capability to tackle the challenges associated with long-term cell culture. We report a marine derived oxidized alginate, alginate dialdehyde (ADA), and gelatin (GEL) system (ADA-GEL), which is cross-linked via ionic (Ca2+) and enzymatic (microbial transglutaminase, mTG) interaction to form dually cross-linked hydrogels. The cross-linking approach allowed us to tailor the stiffness of the hydrogels in a wide range (from <5 to 120 kPa), without altering the initial ADA and GEL hydrogel chemistry. It was possible to control the degradation behavior of the hydrogels to be stable for up to 30 days of incubation. Increasing concentrations of mTG cross-linker solutions allowed us to tune the degradation behavior of the ADA-GEL hydrogels from fast (<7 days) to moderate (14 days) and slow (>30 days) degradation kinetics. The cytocompatibility of mTG cross-linked ADA-GEL was assessed using NIH-3T3 fibroblasts and ATDC-5 mouse teratocarcinoma cells. Both cell types showed highly increased cellular attachment on mTG cross-linked ADA-GEL in comparison to Ca2+ cross-linked hydrogels. In addition, ATDC-5 cells showed a higher proliferation on mTG cross-linked ADA-GEL hydrogels in comparison to tissue culture polystyrene control substrates. Further, the attachment of human umbilical vein endothelial cells (HUVEC) on ADA-GEL (+) mTG was confirmed, proving the suitability of mTG+Ca2+ cross-linked ADA-GEL for several cell types. Summarizing, a promising platform to control the properties of ADA-GEL hydrogels is presented, with the potential to be applied in long-term cell culture investigations such as cartilage, bone, and blood-vessel engineering, as well as for biofabrication.
Collapse
Affiliation(s)
- Thomas Distler
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany
| | - Kilian McDonald
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany
| | - Susanne Heid
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany
| | - Emine Karakaya
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany
| | - Rainer Detsch
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstr. 6, 91058 Erlangen, Germany
| |
Collapse
|
46
|
Al Matari N, Deeb G, Mshiek H, Sinjab A, Kadara H, Abou-Kheir W, Mhanna R. Anti-Tumor Effects of Biomimetic Sulfated Glycosaminoglycans on Lung Adenocarcinoma Cells in 2D and 3D In Vitro Models. Molecules 2020; 25:2595. [PMID: 32503108 PMCID: PMC7321182 DOI: 10.3390/molecules25112595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
Lung cancer development relies on cell proliferation and migration, which in turn requires interaction with extracellular matrix (ECM) components such as glycosaminoglycans (GAGs). The mechanisms through which GAGs regulate cancer cell functions are not fully understood but they are, in part, mediated by controlled interactions with cytokines and growth factors (GFs). In order to mechanistically understand the effect of the degree of sulfation (DS) of GAGs on lung adenocarcinoma (LUAD) cells, we synthesized sulfated alginate (AlgSulf) as sulfated GAG mimics with DS = 0.0, 0.8, 2.0, and 2.7. Human (H1792) and mouse (MDA-F471) LUAD cell lines were treated with AlgSulf of various DSs at two concentrations 10 and 100 µg/mL and their anti-tumor properties were assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), trypan blue exclusion, and wound healing assays for 2D models and sphere formation assay for the 3D model. The proliferation and number of live MDA-F471 cells at the concentration of 100 µg/mL decreased significantly with the increase in the DS of biomimetic GAGs. In addition, the increase in the DS of biomimetic GAGs decreased cell migration (p < 0.001 for DS = 2.0 and 2.7 compared to control) and decreased the diameter and number of spheres formed (p < 0.001). The increased DS of biomimetic GAGs attenuated the expression of cancer stem cell (CSC)/progenitor markers in the 3D cultures. In conclusion, GAG-mimetic AlgSulf with increased DS exhibit enhanced anti-proliferative and migratory properties while also reducing growth of KRAS-mutant LUAD spheres in vitro. We suggest that these anti-tumor effects by GAG-mimetic AlgSulf are possibly due to differential binding to GFs and consequential decreased cell stemness. AlgSulf may be suitable for applications in cancer therapy after further in vivo validation.
Collapse
Affiliation(s)
- Nada Al Matari
- Department of Biomedical Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (N.A.M.); (G.D.)
| | - George Deeb
- Department of Biomedical Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (N.A.M.); (G.D.)
| | - Hiba Mshiek
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (H.K.)
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (H.K.)
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Rami Mhanna
- Department of Biomedical Engineering, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (N.A.M.); (G.D.)
| |
Collapse
|
47
|
Alginate Sulfate Substrates Control Growth Factor Binding and Growth of Primary Neurons: Toward Engineered 3D Neural Networks. ACTA ACUST UNITED AC 2020; 4:e2000047. [DOI: 10.1002/adbi.202000047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/29/2020] [Indexed: 12/27/2022]
|
48
|
Akaraphutiporn E, Sunaga T, Bwalya EC, Echigo R, Okumura M. Alterations in characteristics of canine articular chondrocytes in non-passaged long-term monolayer culture: Matter of differentiation, dedifferentiation and redifferentiation. J Vet Med Sci 2020; 82:793-803. [PMID: 32350166 PMCID: PMC7324834 DOI: 10.1292/jvms.20-0118] [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] [Indexed: 01/16/2023] Open
Abstract
This study investigated the effects of culture time on phenotype stability of canine
articular chondrocytes (CACs) in non-passaged long-term monolayer culture. Third passage
(P3) CACs isolated from four cartilage samples were seeded at three different initial
seeding densities (0.2 × 104, 1.0 × 104 and 5.0 × 104
cells/cm2) and maintained in monolayer condition up to 8 weeks without
undergoing subculture after confluence. The characteristic changes of chondrocytes during
the culture period were evaluated based on the cell morphology, cell proliferation,
glycosaminoglycans (GAGs) content, DNA quantification, mRNA expression and ultrastructure
of chondrocytes. Chondrocytes maintained under post-confluence condition exhibited a
capability to grow and proliferate up to 4 weeks. Alcian blue staining and
Dimethylmethylene blue (DMMB) assay revealed that the extracellular matrix (ECM) synthesis
was increased in a time-dependent manner from 2 to 8 weeks. The chondrocyte mRNA
expression profile was dramatically affected by prolonged culture time, with a significant
downregulation of collagen type I, whereas the expression of
collagen type II, aggrecan, Sox9 and
matrix metalloproteinase 13 (MMP-13) were significantly upregulated. In
addition, transmission electron microscopy (TEM) result indicated dilation of rough
endoplasmic reticulum (RER) in these long-term monolayer cultured chondrocytes. These
findings demonstrate that the chondrocytes phenotype could be partially redifferentiated
through the spontaneous redifferentiation process in long-term cultures using standard
culture medium without the addition of chondrogenic supplements or tissue-culture
scaffolds.
Collapse
Affiliation(s)
- Ekkapol Akaraphutiporn
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Takafumi Sunaga
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Eugene C Bwalya
- Department of Clinical Studies, Samora Machel School of Veterinary Medicine, University of Zambia, Lusaka 10101, Zambia
| | - Ryosuke Echigo
- Veterinary Medical Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Masahiro Okumura
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
49
|
Szabó L, Gerber-Lemaire S, Wandrey C. Strategies to Functionalize the Anionic Biopolymer Na-Alginate without Restricting Its Polyelectrolyte Properties. Polymers (Basel) 2020; 12:E919. [PMID: 32326625 PMCID: PMC7240516 DOI: 10.3390/polym12040919] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/09/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022] Open
Abstract
The natural anionic polyelectrolyte alginate and its derivatives are of particular interest for pharmaceutical and biomedical applications. Most interesting for such applications are alginate hydrogels, which can be processed into various shapes, self-standing or at surfaces. Increasing efforts are underway to functionalize the alginate macromolecules prior to hydrogel formation in order to overcome the shortcomings of purely ionically cross-linked alginate hydrogels that are hindering the progress of several sophisticated biomedical applications. Particularly promising are derivatives of alginate, which allow simultaneous ionic and covalent cross-linking to improve the physical properties and add biological activity to the hydrogel. This review will report recent progress in alginate modification and functionalization with special focus on synthesis procedures, which completely conserve the ionic functionality of the carboxyl groups along the backbone. Recent advances in analytical techniques and instrumentation supported the goal-directed modification and functionalization.
Collapse
Affiliation(s)
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC SCI-SB-SG, Station 6, CH-1015 Lausanne, Switzerland; (L.S.); (C.W.)
| | | |
Collapse
|
50
|
Cao C, Zhang Y, Ye Y, Sun T. Effects of cell phenotype and seeding density on the chondrogenic capacity of human osteoarthritic chondrocytes in type I collagen scaffolds. J Orthop Surg Res 2020; 15:120. [PMID: 32228637 PMCID: PMC7104498 DOI: 10.1186/s13018-020-01617-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 02/27/2020] [Indexed: 01/01/2023] Open
Abstract
Objective Matrix-associated autologous chondrocyte implantation (MACI) achieves good clinical efficacy in young patients with focal cartilage injury; however, phenotypic de-differentiation of chondrocytes cultured in monolayer and the treatment of older OA patients are still challenges in the field of cartilage tissue engineering. This study aimed to assess the in vitro re-differentiation potential and in vivo chondrogenic capacity of human OA chondrocytes inoculated into collagen I scaffolds with different cellular phenotypes and seeding densities. Methods OA chondrocytes and articular chondrocyte-laden scaffolds were cultured over 2 weeks in in vitro. Reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) and histological staining were used to detect the mRNA expression profiles and extracellular matrix secretion of chondrocyte-specific markers. OA chondrocyte-laden collagen I scaffolds with different cellular phenotypes, and seeding densities were implanted into SCID mice over 4 weeks to evaluate the chondrogenic capacity in vivo. Results Increased COL2a1, ACAN, COMP, SOX9, and BMP2 expression levels and decreased COL1a1, VCAN, MMP13, and ADAMTS5 amounts were observed in OA chondrocytes seeded in collagen I scaffolds; Implantation of phenotypically superior OA chondrocytes in collagen I scaffolds at high density could improve the chondrogenic capacity of human OA chondrocytes, as confirmed by RT-qPCR assessed gene expression patterns in vitro and histological evaluation in vivo. Conclusions Freshly isolated chondrocytes from OA patients could be a source of replacement for articular chondrocytes being commonly used in MACI. Implantation of phenotypically superior OA chondrocytes in collagen I scaffolds at high density could be a promising tool for the treatment of elderly OA patients.
Collapse
Affiliation(s)
- Chenxi Cao
- Arthritis Clinic and Research Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China.,Institute of Sports Medicine, Peking University Third Hospital, Beijing, 100044, People's Republic of China
| | - Yujun Zhang
- The Institute of Clinical Molecular Biology and the Central Lab, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Yanqi Ye
- Arthritis Clinic and Research Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Tiezheng Sun
- Arthritis Clinic and Research Center, Peking University People's Hospital, Beijing, 100044, People's Republic of China.
| |
Collapse
|